CA2940742A1 - Regulators of frataxin - Google Patents

Regulators of frataxin Download PDF

Info

Publication number
CA2940742A1
CA2940742A1 CA2940742A CA2940742A CA2940742A1 CA 2940742 A1 CA2940742 A1 CA 2940742A1 CA 2940742 A CA2940742 A CA 2940742A CA 2940742 A CA2940742 A CA 2940742A CA 2940742 A1 CA2940742 A1 CA 2940742A1
Authority
CA
Canada
Prior art keywords
bet
frataxin
pharmaceutically acceptable
acceptable salt
optionally substituted
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
CA2940742A
Other languages
French (fr)
Inventor
Eugene Lvovich Piatnitski Chekler
Leslie Anthony DAKIN
Lyn Howard Jones
Joseph Fouad NABHAN
Mathew Tyler PLETCHER
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Pfizer Inc
Original Assignee
Pfizer Inc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Pfizer Inc filed Critical Pfizer Inc
Publication of CA2940742A1 publication Critical patent/CA2940742A1/en
Abandoned legal-status Critical Current

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/55Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having seven-membered rings, e.g. azelastine, pentylenetetrazole
    • A61K31/551Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having seven-membered rings, e.g. azelastine, pentylenetetrazole having two nitrogen atoms, e.g. dilazep
    • A61K31/55131,4-Benzodiazepines, e.g. diazepam or clozapine
    • A61K31/55171,4-Benzodiazepines, e.g. diazepam or clozapine condensed with five-membered rings having nitrogen as a ring hetero atom, e.g. imidazobenzodiazepines, triazolam
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/47Quinolines; Isoquinolines
    • A61K31/4709Non-condensed quinolines and containing further heterocyclic rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/47Quinolines; Isoquinolines
    • A61K31/4738Quinolines; Isoquinolines ortho- or peri-condensed with heterocyclic ring systems
    • A61K31/4745Quinolines; Isoquinolines ortho- or peri-condensed with heterocyclic ring systems condensed with ring systems having nitrogen as a ring hetero atom, e.g. phenantrolines
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/535Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with at least one nitrogen and one oxygen as the ring hetero atoms, e.g. 1,2-oxazines
    • A61K31/53751,4-Oxazines, e.g. morpholine
    • A61K31/53771,4-Oxazines, e.g. morpholine not condensed and containing further heterocyclic rings, e.g. timolol
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/55Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having seven-membered rings, e.g. azelastine, pentylenetetrazole
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P21/00Drugs for disorders of the muscular or neuromuscular system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/14Drugs for disorders of the nervous system for treating abnormal movements, e.g. chorea, dyskinesia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/28Drugs for disorders of the nervous system for treating neurodegenerative disorders of the central nervous system, e.g. nootropic agents, cognition enhancers, drugs for treating Alzheimer's disease or other forms of dementia

Abstract

This invention relates to a use of an effective amount of a BET- family bromodomain inhibitor, or a pharmaceutically acceptable salt thereof, for increasing the level of frataxin, or for increasing the expression of frataxin. This invention also relates to a use of of a BET- family bromodomain inhibitor, or a pharmaceutically acceptable salt thereof, or a composition thereof for treating a condition or a disease associated with decreased levels or activity of frataxin, including Friedreich's ataxia

Description

, PC72246 REGULATORS OF FRATAXIN
FIELD
The present invention relates to regulators of frataxin and, in particular, to regulating the level of frataxin or the expression of frataxin using a BET-family bromodomain inhibitor, or a pharmaceutically acceptable salt thereof.
Associated pharmaceutical uses and pharmaceutical compositions are also described.
BACKGROUND
Friedreich's ataxia (also called FA or FRDA) is a rare recessive inherited disease characterized by progressive damage to the nervous system and movement problems.
The first symptoms of the condition are usually gait ataxia or difficultly walking, but overtime the symptoms worsen and spread resulting in degeneration of the spinal cord, peripheral nerves and cerebellum. The disorder also causes problems in the spine and loss of tendon reflex. Most people with Friedreich's ataxia develop scoliosis (a curving of the spine to one side), which often requires surgical intervention for treatment.
Dysarthria (slowness and slurring of speech) may also develop as well as hearing and vision loss. Other symptoms that may occur include chest pain, shortness of breath, and heart palpitations. These symptoms are the result of various forms of heart disease that often accompany Friedreich's ataxia, such as hypertrophic cardiomyopathy (enlargement of the heart), myocardial fibrosis (formation of fiber-like material in the muscles of the heart), and cardiac failure. Heart rhythm abnormalities such as tachycardia (fast heart rate) and heart block (impaired conduction of cardiac impulses within the heart) are also common. About 20 percent of people with Friedreich's ataxia develop carbohydrate intolerance and 10 percent develop diabetes. Symptoms typically begin between the ages of 5 and 15 years and generally within 10 to 20 years after the appearance of the first symptoms the patient is confined to a wheelchair and in later stages of the disease individuals may become completely incapacitated.
Friedreich's ataxia can lead to early death, often as a result of associated heart disease.
Friedreich's ataxia is caused by reduced expression of frataxin (FXN) as a result of inheriting two copies of the mutated FXN gene which disrupts the normal production , PC72246
2 of frataxin. Frataxin is found in the mitochondria and research suggests that without a normal level of frataxin, certain cells in the body (especially peripheral nerve, spinal cord, brain and heart muscle cells) cannot effectively produce energy and it is further hypothesized that these cells experience oxidative stress due to the buildup of toxic byproducts. Frataxin is initially synthesized in vivo as a precursor polypeptide which is in turn directed to the mitochondrial matrix where it is proteolytically cleaved by mitochondrial processing peptidase to the mature form of frataxin via a processing intermediate frataxin ("The in vitro mitochondrial two-step maturation of human frataxin."
Stephane Schmucker et al, Human Molecular Genetics, 2008, vol 17 no 22, p3521-3531). It is also thought that reduced expression of frataxin results in the nervous system, heart, and pancreas being particularly susceptible to damage from free radicals.
As with many degenerative diseases of the nervous system, there is currently no known cure or effective treatment for Friedreich's ataxia. To date, the only therapy options remain symptomatic treatment to help individuals maintain optimal functioning for as long as possible. For example, prescription of medication to treat diabetes or heart problems, use of surgery or braces or physical therapy to treat scoliosis and other skeletal problems.
There remains a need for new small molecule approaches for the regulation of frataxin, and which may therefore be used in the treatment of Friedreich's ataxia or related conditions, diseases or disorders associated with the regulation of frataxin.
SUMMARY
This invention relates to a method of regulating the expression of frataxin in a cell, comprising contacting the cell with an effective amount of a BET- family bromodomain inhibitor, or a pharmaceutically acceptable salt thereof.
This invention also relates to a method of increasing the expression of frataxin in a cell, comprising contacting the cell with an effective amount of a BET-family bromodomain inhibitor, or a pharmaceutically acceptable salt thereof.

, PC72246
3 This invention further relates to a use of an effective amount of a BET-family bromodomain inhibitor, or a pharmaceutically acceptable salt thereof, for regulating the expression of frataxin.
This invention also relates to a use of an effective amount of a BET- family bromodomain inhibitor, or a pharmaceutically acceptable salt thereof, for increasing the expression of frataxin.
This invention also relates to a use of an effective amount of a BET- family bromodomain inhibitor, or a pharmaceutically acceptable salt thereof, for increasing the expression of frataxin in a cell or a subject which cell or subject has decreased frataxin expression when compared to a healthy control cell or healthy subject.
This invention also relates to a use of an effective amount of a BET- family bromodomain inhibitor, or a pharmaceutically acceptable salt thereof, for increasing the level of frataxin.
This invention also relates to a use of an effective amount of a BET- family bromodomain inhibitor, or a pharmaceutically acceptable salt thereof, for increasing the level of frataxin in a cell or a subject which cell or subject has decreased level of frataxin when compared to a healthy control cell or healthy subject.
This invention also relates to a use of a therapeutically effective amount of a BET- family bromodomain inhibitor, or a pharmaceutically acceptable salt thereof, for treating a condition, disease or disorder, or the symptoms associated with the condition, disease or disorder, which condition, disease or disorder is associated with decreased frataxin expression ma subject in need thereof.
This invention also relates to a use of a therapeutically effective amount of a BET- family bromodomain inhibitor, or a pharmaceutically acceptable salt thereof, for treating a condition, disease or disorder, or the symptoms associated with the condition, disease or disorder, which condition, disease or disorder is associated with a decreased level of frataxin in a subject in need thereof.
This invention also relates to a use of a therapeutically effective amount of a BET- family bromodomain inhibitor, or a pharmaceutically acceptable salt thereof, for treating Friedreich's ataxia in a subject in need thereof.
4 This invention also relates to a BET-family bromodomain inhibitor, or a pharmaceutically acceptable salt thereof, for use in the treatment of a condition, disease or disorder, or the symptoms associated with the condition, disease or disorder, which condition, disease or disorder is associated with decreased frataxin expression.
This invention also relates to a BET-family bromodomain inhibitor, or a pharmaceutically acceptable salt thereof, for use in the treatment of a condition, disease or disorder, or the symptoms associated with the condition, disease or disorder, which condition, disease or disorder is associated with a decreased level of frataxin.
This invention also relates to a BET-family bromodomain inhibitor, or a pharmaceutically acceptable salt thereof, for use in the treatment of Friedreich's ataxia.
This invention also relates to the use of a BET-family bromodomain inhibitor, or a pharmaceutically acceptable salt thereof, in the manufacture of a medicament for the treatment of a condition, disease or disorder, or the symptoms associated with the condition, disease or disorder, which condition, disease or disorder is associated with decreased frataxin expression.
This invention also relates to the use of a BET-family bromodomain inhibitor, or a pharmaceutically acceptable salt thereof, in the manufacture of a medicament for the treatment of a condition, disease or disorder, or the symptoms associated with the condition, disease or disorder, which condition, disease or disorder is associated with a decreased level of frataxin.
This invention also relates to the use of a BET-family bromodomain inhibitor, or a pharmaceutically acceptable salt thereof, in the manufacture of a medicament for the treatment of Friedreich's ataxia.
This invention also relates to a pharmaceutical composition comprising a BET-family bromodomain inhibitor, or a pharmaceutically acceptable salt thereof, for the treatment of a condition, disease or disorder, or the symptoms associated with the condition, disease or disorder, which condition, disease or disorder is associated with decreased frataxin expression.
This invention also relates to a pharmaceutical composition comprising a BET-family bromodomain inhibitor, or a pharmaceutically acceptable salt thereof, for the treatment of a condition, disease or disorder, or the symptoms associated with the condition, disease or disorder, which condition, disease or disorder is associated with a decreased level of frataxin.
This invention also relates to a pharmaceutical composition comprising a BET-family bromodomain inhibitor, or a pharmaceutically acceptable salt thereof, for the
5 treatment of Friedreich's ataxia.
BRIEF DESCRIPTION OF FIGURES
Figure 1 shows that BET-family bromodomain inhibitors IBET-762 and JQ-1(+) increased levels of both intermediate frataxin (bands "i") and mature frataxin (bands "m") at 72 hrs by Western blot (two exposures shown) in FRDA GM15850 cells.
Figure 2 shows the cytotoxic effects of treatment of the FRDA GM15850 cells with BET-family bromodomain inhibitors IBET-762 and JQ-1(+) (protein concentrations (mg/ml) for the resultant lysates).
Figure 3 shows that a minimally cyctotoxic concentration of BET-family bromodomain inhibitor JQ-1(+) increased levels of both intermediate frataxin (bands "i") and mature frataxin (bands "m") at 72 hrs by Western blot (two exposures shown) in FRDA GM15850 cells. Results from DMSO treated AG14725 healthy cells control are also shown.
Figure 4 shows pixel densitometry quantification (base line corrected) of immunopositive bands corresponding to mature frataxin from the Western blot of Figure 3. An approximately 1.5 fold increase was observed at 25nM and 50nM treatment with BET-family bromodomain inhibitor JQ-1(+) when compared to DMSO vehicle treated controls (p<0.05).
Figure 5 shows a similar cellular toxicity profile (LD50) for both FRDA

lymphoblast cells and healthy AG14725 upon treatment with BET-family bromodomain inhibitor JQ-1(+), suggesting that a lower level of frataxin does not sensitize cells to toxicitiy by the BET-family bromodomain inhibitor JQ-1(+).
Figure 6 shows that BET-family bromodomain inhibitors with a variety of chemical scaffolds (compounds 1 ¨ 6), increased levels of both intermediate frataxin (bands "i") and mature frataxin (bands "m") at 72 hrs by Western blot (two exposures shown) in FRDA GM15850 cells.
6 Figure 7 shows that BRD4 knockdown using the BRD4 siRNA smartpool or treatment with BET-family bromodomain inhibitor JQ-1(+) increased levels of both intermediate frataxin (bands "i") and mature frataxin (bands "m") at 72 hrs by Western blot (two exposures shown) in FRDA GM03665 fibroblasts.
Figure 8 shows that knockdown of other BET-family proteins using siRNA
smartpools also resulted in increased levels of both intermediate frataxin (bands "i") and mature frataxin (bands "m") at 72 hrs by Western blot (two exposures shown) in FRDA
GM03665 fibroblasts.
DETAILED DESCRIPTION
The present invention relates to regulating the level of frataxin or the expression of frataxin using a BET-family bromodomain inhibitor, or a pharmaceutically acceptable salt thereof. The present invention also relates to novel pharmaceutical uses for associated treatment, utilising a BET-family bromodomain inhibitor, or a pharmaceutically acceptable salt thereof.
Uses and Pharmaceutical Uses for Treatment This invention relates to a use of an effective amount of a BET- family bromodomain inhibitor, or a pharmaceutically acceptable salt thereof, for regulating the expression of frataxin.
This invention also relates to a use of an effective amount of a BET- family bromodomain inhibitor, or a pharmaceutically acceptable salt thereof, for increasing the expression of frataxin.
This invention also relates to a use of an effective amount of a BET- family bromodomain inhibitor, or a pharmaceutically acceptable salt thereof, for increasing the expression of frataxin in a cell or a subject which cell or subject has decreased frataxin expression when compared to a healthy control cell or healthy subject.
This invention also relates to a use of an effective amount of a BET- family bromodomain inhibitor, or a pharmaceutically acceptable salt thereof,for increasing the level of frataxin.
7 This invention also relates to a use of an effective amount of a BET- family bromodomain inhibitor, or a pharmaceutically acceptable salt thereof, for increasing the level of frataxin in a cell or a subject which cell or subject has decreased level of frataxin when compared to a healthy control cell or healthy subject.
This invention also relates to a use of a therapeutically effective amount of a BET- family bromodomain inhibitor, or a pharmaceutically acceptable salt thereof, for treating a condition, disease or disorder, or the symptoms associated with the condition, disease or disorder, which condition, disease or disorder is associated with decreased frataxin expression, in a subject in need thereof.
This invention also relates to a use of a therapeutically effective amount of a BET- family bromodomain inhibitor, or a pharmaceutically acceptable salt thereof, for treating a condition, disease or disorder, or the symptoms associated with the condition, disease or disorder, which condition, disease or disorder is associated with a decreased level of frataxin, in a subject in need thereof.
This invention also relates to a use of a therapeutically effective amount of a BET- family bromodomain inhibitor, or a pharmaceutically acceptable salt thereof, for treating Friedreich's ataxia.
This invention also relates to a BET-family bromodomain inhibitor, or a pharmaceutically acceptable salt thereof, for use in the treatment of a condition, disease or disorder, or the symptoms associated with the condition, disease or disorder, which condition, disease or disorder is associated with decreased frataxin expression.
This invention also relates to a BET-family bromodomain inhibitor, or a pharmaceutically acceptable salt thereof, for use in the treatment of a condition, disease or disorder, or the symptoms associated with the condition, disease or disorder, which condition, disease or disorder is associated with a decreased level of frataxin.
This invention also relates to a BET-family bromodomain inhibitor, or a pharmaceutically acceptable salt thereof, for use in the treatment of Friedreich's ataxia.
This invention also relates to the use of a BET-family bromodomain inhibitor, or a pharmaceutically acceptable salt thereof, in the manufacture of a medicament for the treatment of a condition, disease or disorder, or the symptoms associated with the
8 condition, disease or disorder, which condition, disease or disorder is associated with decreased frataxin expression.
This invention also relates to the use of a BET-family bromodomain inhibitor, or a pharmaceutically acceptable salt thereof, in the manufacture of a medicament for the treatment of a condition, disease or disorder, or the symptoms associated with the condition, disease or disorder, which condition, disease or disorder is associated with a decreased level of frataxin.
This invention also relates to the use of a BET-family bromodomain inhibitor, or a pharmaceutically acceptable salt thereof, in the manufacture of a medicament for the treatment of Friedreich's ataxia.
This invention also relates to a pharmaceutical composition comprising a BET-family bromodomain inhibitor, or a pharmaceutically acceptable salt thereof, for the treatment of a condition, disease or disorder, or the symptoms associated with the condition, disease or disorder, which condition, disease or disorder is associated with decreased frataxin expression.
This invention also relates to a pharmaceutical composition comprising a BET-family bromodomain inhibitor, or a pharmaceutically acceptable salt thereof, for the treatment of a condition, disease or disorder, or the symptoms associated with the condition, disease or disorder, which condition, disease or disorder is associated with a decreased level of frataxin.
This invention also relates to a pharmaceutical composition comprising a BET-family bromodomain inhibitor, or a pharmaceutically acceptable salt thereof, for the treatment of Friedreich's ataxia.
In one embodiment, as used herein, the terms "treat" or "treating" may refer to partially or completely alleviating, inhibiting, ameliorating and/or relieving a condition, disease or disorder, or the symptoms associated with the condition, disease or disorder from which a patient or subject has been diagnosed as suffering or from which a patient or subject is suspected to suffer.
In another embodiment, as used herein, the terms "treat" or "treating" may also refer to slowing of the advancement of a condition, disease or disorder, or the symptoms associated with the condition, disease or disorder from which a patient or
9 subject has been diagnosed as suffering or from which a patient or subject is suspected to suffer.
In another embodiment, the terms "treat" or "treating" may also refer to regression of the condition, disease or disorder, including regression of the symptoms associated with the condition, disease or disorder, from which a patient or subject has been diagnosed as suffering or from which a patient or subject is suspected to suffer.
In another embodiment, the terms "treat" or "treating" may also refer to the prevention of a condition, disease or disorder, or the symptoms associated with the condition, disease or disorder from which a patient or subject has been diagnosed as suffering or from which a patient or subject is suspected to suffer, or from which a patient or subject is at high risk of suffering.
In another embodiment, as used herein, the terms "treat" or "treating" may also refer to the prevention of the advancement of a condition, disease or disorder, or the symptoms associated with the condition, disease or disorder, from which a patient or subject has been diagnosed as suffering or from which a patient or subject is suspected to suffer.
The term "effective amount" as used herein refers to an amount of an agent that elicits the desired biological activity or effect.
The term "therapeutically effective" as used herein refers to an agent or an amount of an agent that elicits a desirable biological therapeutic activity or desirable therapeutic effect.
As used herein, except when noted, the terms "subject" or "patient" are used interchangeably and refer to mammals. The term "mammals" may include humans and non-human primates, as well as experimental animals such as rabbits, rats, and mice.
In one embodiment the subject or patient may be a human. In another embodiment the subject or patient may be a male human. In another embodiment the subject or patient may be a female human. Accordingly, the term "subject" or "patient" as used herein may mean any mammalian patient or subject to which the compounds useful for the present invention may be administered.
In one embodiment of the present invention, routine screening methods are used to either identify and/or diagnose patients for treatment according to the invention; to identify the existence of a specific condition, disease or disorder which is associated with decreased expression of frataxin; to identify the existence of a specific condition, disease or disorder which is associated with a decreased level of frataxin; to determine risk factors associated with a targeted or suspected condition, disease or disorder; or to 5 determine the status of an existing condition, disease or disorder in the subject. These routine screening methods include, but are not limited to, for example, conventional work-ups to determine risk factors that may be associated with the targeted or suspected condition, disease or disorder; conventional genetic screening methods and the like. These and other routine methods allow identification of patients in need of
10 therapy using the compounds of the present invention.
In one embodiment the subject is a patient diagnosed with a condition, disease or disorder, or symptoms associated with a condition, disease or disorder, which condition, disease or disorder is associated with decreased expression of frataxin. In one embodiment the subject is a patient diagnosed with a condition, disease or disorder, or symptoms associated with a condition, disease or disorder, which condition, disease or disorder is associated with decreased expression of frataxin when compared to a healthy subject or patient. In one embodiment the subject is a patient diagnosed with a condition, disease or disorder, or symptoms associated with a condition, disease or disorder, which condition, disease or disorder is associated with a decreased level of frataxin. In one embodiment the subject is a patient diagnosed with a condition, disease or disorder, or symptoms associated with a condition, disease or disorder, which condition, disease or disorder is associated with a decreased level of frataxin when compared to a healthy subject or patient. In one embodiment the subject is a patient diagnosed with a condition, disease or disorder, which condition, disease or disorder is associated with decreased frataxin expression, but which patient or subject is asymptomatic. In one embodiment the subject is a patient diagnosed with a condition, disease or disorder, which condition, disease or disorder is associated with a decreased level of frataxin, but which patient or subject is asymptomatic. In one embodiment the subject is a patient diagnosed with Friedreich's ataxia. In one embodiment the subject is a patient diagnosed with Friedreich's ataxia but is asymptomatic.
In one embodiment the subject may be a patient diagnosed with Friedreich's ataxia with retained reflexes. In one embodiment the subject may be a patient
11 diagnosed with Friedreich's ataxia with retained reflexes but is asymptomatic.
In one embodiment the subject may be a patient diagnosed with late onset Friedreich's ataxia.
In one embodiment the subject may be a patient diagnosed with late onset Friedreich's ataxia but is asymptomatic. In one embodiment the subject may be asymptomatic but has one or more genetic risk factors associated with a condition, disease or disorder, which condition, disease or disorder is associated with decreased frataxin expression.
In one embodiment the subject may be asymptomatic but has one or more genetic risk factors associated with a condition, disease or disorder, which condition, disease or disorder is associated with a decreased level of frataxin. In one embodiment the subject may be asymptomatic but has one or more genetic risk factors associated with Friedreich's ataxia. In one embodiment the subject may be asymptomatic but is judged to be at high risk of developing a condition, disease or disorder associated with decreased frataxin expression. In one embodiment the subject may be asymptomatic but is judged to be at high risk of developing a condition, disease or disorder associated with a decreased level of frataxin.
In one embodiment the subject may be asymptomatic but is judged to be at high risk of developing Friedreich's ataxia. In one embodiment the subject may be asymptomatic but is judged to be a carrier of Friedreich's ataxia.
In one embodiment the patient or subject may be identified as having greater than about 75 unstable homozygous GAA trinucleotide repeat expansions in the intron-1 on both of the alleles in the gene which encodes frataxin. In one embodiment the patient or subject may be identified as having greater than about 75 unstable homozygous GAA trinucleotide repeat expansions in the intron-1 on one of the alleles in the gene which encodes frataxin. In one embodiment the patient or subject may be identified as having between about 75 and about 300 GAA triplet repeat expansions in the intron-1 on both of the alleles in the gene which encodes frataxin. In one embodiment the patient or subject may be identified as having between about 75 and about 300 GAA triplet repeat expansions in the intron-1 on one of the alleles in the gene which encodes frataxin. In one embodiment the patient or subject may be identified as having greater than about 300 GAA triplet repeat expansions in the intron-1 on both of the alleles in the gene which encodes frataxin. In one embodiment the patient or subject may be identified as having greater than about 300 GAA triplet repeat expansions in the intron-1 on one of the alleles in the gene which encodes frataxin. In = PC72246
12 one embodiment the subject or patient may be identified as being compound heterozygous for a GAA repeat expansion in the intron-1 of the gene which encodes frataxin and a point mutation or deletion.
In one embodiment the "patient" or "subject" with the condition, disease or disorder, or the symptoms associated with the condition, disease or disorder, may have either decreased frataxin expression or a decreased level of frataxin such that the residual cellular level of frataxin is from about 2% to about 30%; or from about 5% to about 25%; or from about 10% to about 20% of the residual cellular level of frataxin of a healthy counterpart.
As used herein the term "Friedreich's ataxia" shall be taken to mean an autosomal recessive congenital ataxia caused by a mutation(s) in the gene which encodes frataxin.
In one embodiment, a subject or patient who has been identified as having greater than about 300 unstable homozygous GAA trinucleotide repeat expansions in the intron-1 on both of the alleles in the gene which encodes frataxin may be treated for Friedreich's ataxia in accordance with the invention. In one embodiment, a subject or patient who has been identified as having late stage on-set of Friedreich's ataxia may be treated for Friedreich's ataxia in accordance with the invention. In one embodiment, a subject or patient who has greater than from about 75 to about 300 unstable homozygous GAA trinucleotide repeat expansion in the intron-1 on both of the alleles in the gene which encodes frataxin may be treated for late stage on-set of Friedreich's ataxia in accordance with the invention. In one embodiment a subject or patient who has been identified as a heterozygous carrier of Friedreich's ataxia may be treated for Friedreich's ataxia in accordance with the invention. In one embodiment, a subject or patient who has greater than about 75 unstable homozygous GAA trinucleotide repeat expansion in the intron-1 on one of the alleles in the gene which encodes frataxin may be treated as a carrier of Friedreich's ataxia in accordance with the invention. In one embodiment, the subject or patient may be identified as being compound heterozygous for a GAA repeat expansion in the intron-1 of the gene which encodes frataxin and a point mutation or deletion.
In one embodiment the symptoms associated with the condition, disease or disorder may include muscle weakness in the arms and legs, loss of co-ordination, loss
13 of deep tendon reflexes, loss of extensor plantar responses, loss of vibratory and proprioceptive sensation, vision impairment, involuntary and / or rapid eye movements, hearing impairment, slurred speech, curvature of the spine (scoliosis), high plantar arches (pes cavus deformity of the foot), carbohydrate intolerance, diabetes mellitus and heart disorders (including but not limited to atrial fibrillation, tachycardia (fast heart rate), hypertrophic card iomyopathy, card iomegaly, symmetrical hypertrophy, heart murmurs, and heart conduction defects).
As used herein, the term "mammal" may include, but is not limited to, mammals such as a human, a non-human primate, canine, feline, bovine, ovine, porcine, murine, or other veterinary or laboratory mammals such as rabbits, rats, and mice.
Those skilled in the art recognize that a therapy which reduces the severity of pathology in one species of mammal is predictive of the effect of the therapy on another species of mammal.
As used herein, the term "modulate" shall be taken to encompass either a decrease or an increase in the expression, level or activity depending on the target molecule.
As used herein the term "regulating the expression of" shall be taken to mean either an intervention which results in a decrease in the expression of the target molecule, or an intervention which results in an increase in the expression of the target molecule. In one embodiment the regulation is an intervention which results in an "increase" or "up-regulation", which terms are used herein interchangeably, and which, as used herein, shall be taken to mean resulting in, either directly or indirectly, an increase in the level of the target molcule expressed.
As used herein, the term "increasing the expression of frataxin" shall be taken to mean that the cellular expression of frataxin precursor has, either directly or indirectly, been increased when compared to the cellular expression of frataxin precursor prior to treatment in accordance with the invention. Without wishing to be bound by theory, it is thought that affecting an increase in the expression of frataxin precursor will result initially in an increase in the level of frataxin precursor which will in turn lead to an increase in the level of one or both of intermediate frataxin and mature frataxin as the frataxin precursor is further metabolized. Furthermore, without wishing to be bound by theory, in one embodiment it is thought that the expression of frataxin precursor is = ' ' PC72246
14 directly increased by modulation of the epigenetic state of the gene which encodes frataxin for example by either direct or indirect downstream modulation of P-TEFb and RNA polymerase II; by modulation of histone methylation; by modulation of transcriptional pause release; by relieving repression; or by modulating the methylation of CPG's in the DNA. Alternatively it is possible that the expression of frataxin precursor is directly increased by the modulation of the epigenetic state via relieving heterochromatinisation. In another embodiment, it is possible that the expression of frataxin precursor is modulated indirectly for example by inhibiting the expression of a negative regulator of frataxin, for example the negative anti-sense oligo repressor of expression FAST-1, or by inducing the expression of a positive regulator of frataxin. For example, without wishing to be bound by theory, epigenetic silencing in Friedreich's ataxia may be associated with depletion of CTCF (CCCTC-binding factor) and antisense transcription.
As used herein, the term "increasing the level of frataxin" shall be taken to mean that the level of frataxin, including frataxin precursor, intermediate frataxin and / or mature frataxin, in a cell, subject or patient has, either directly or indirectly, been increased when compared to the level of frataxin, including frataxin precursor, intermediate frataxin and / or mature frataxin, in a cell subject or patient prior to treatment in accordance with the invention. Without wishing to be bound by theory, one means for directly increasing the level of frataxin, including frataxin precursor, intermediate frataxin and / or mature frataxin, in a cell, subject or patient is to increase the expression of frataxin. Alternatively, without wishing to be bound by theory, means for indirectly increasing the level of frataxin, including frataxin precursor, intermediate frataxin and / or mature frataxin, in a cell, subject or patient may include either increasing the stability of frataxin mRNA and / or decreasing the rate of degradation of frataxin, including frataxin precursor, intermediate frataxin, and / or mature frataxin.
In one embodiment the level of frataxin, including frataxin precursor, intermediate frataxin and / or mature frataxin, may be increased by at least about 1.5 fold when compared to the level of frataxin, including frataxin precursor, intermediate and / or mature frataxin in the cell, subject or patient prior to treatment in accordance with the invention. In one embodiment the level of frataxin, including frataxin precursor, intermediate frataxin and / or mature frataxin, may be increased by at least about 2 fold = i= PC72246 when compared to the level of frataxin, including frataxin precursor, intermediate frataxin and / or mature frataxin, in the cell, subject or patient prior to treatment in accordance with the invention. In one embodiment the level of frataxin, including frataxin precursor, intermediate frataxin and / or mature frataxin, may be increased by at least about 5 fold when compared to the level of frataxin, including frataxin precursor, intermediate frataxin and / or mature frataxin, in the cell, subject or patient prior to treatment in accordance with the invention. In one embodiment, the level of frataxin, including frataxin precursor, intermediate frataxin and / or mature frataxin, may be measured as the steady state level after a period of treatment.

In one embodiment the level of frataxin, including frataxin precursor, intermediate frataxin and / or mature frataxin, in the cell, subject or patient treated in accordance with the invention may be increased to at least about 10% of the level of frataxin, including frataxin precursor, intermediate frataxin and / or mature frataxin, in a healthy cell or a healthy subject.
In another embodiment the level of frataxin, including frataxin
15 precursor, intermediate frataxin and / or mature frataxin, in the cell, subject or patient treated in accordance with the invention may be increased to at least about 20% of the level of frataxin, including frataxin precursor, intermediate frataxin and /
or mature frataxin, in a healthy cell or a healthy subject. In another embodiment the level of frataxin, including frataxin precursor, intermediate frataxin and / or mature frataxin, in the cell, subject or patient treated in accordance with the invention may be increased to at least about 30% of the level of frataxin, including frataxin precursor, intermediate frataxin and / or mature frataxin, in a healthy cell or a healthy subject. In another embodiment the level of frataxin, including frataxin precursor, intermediate frataxin and /
or mature frataxin, in the cell, subject or patient treated in accordance with the invention may be increased to at least about 40% of the level of frataxin, including frataxin precursor, intermediate frataxin and / or mature frataxin, in a healthy cell or a healthy subject. In another embodiment the level of frataxin, including frataxin precursor, intermediate frataxin and / or mature frataxin, in the cell, subject or patient treated in accordance with the invention may be increased to at least about 50% of the level of frataxin, including frataxin precursor, intermediate frataxin and / or mature frataxin, in a healthy cell or a healthy subject. In another embodiment the level of frataxin, including frataxin precursor, intermediate frataxin and / or mature frataxin, in the cell, subject or patient treated in accordance with the invention may be increased to at least about 75%

' PC72246
16 of the level of frataxin, including frataxin precursor, intermediate frataxin and / or mature frataxin, in a healthy cell or a healthy subject. In another embodiment the level of frataxin, including frataxin precursor, intermediate frataxin and / or mature frataxin, in the cell, subject or patient treated in accordance with the invention may be increased to at least about 100% of the level of frataxin, including frataxin precursor, intermediate frataxin and / or mature frataxin, in a healthy cell or a healthy subject.
In one embodiment, the level of frataxin, including frataxin precursor, intermediate frataxin and / or mature frataxin, in the cell, subject or patient treated in accordance with the invention may be increased to about that of a heterozygous carrier of Friedreich's ataxia. In another embodiment, the level of frataxin, including frataxin precursor, intermediate frataxin and/or mature frataxin, in the cell, subject or patient treated in accordance with the invention may be increased to more than about that of a heterozygous carrier of Friedreich's ataxia. In another embodiment, the level of frataxin, including frataxin precursor, intermediate frataxin and/or mature frataxin, in the cell, subject or patient treated in accordance with the invention may be increased to be about the same as that of a healthy cell or subject. In one embodiment, the level of frataxin, including frataxin precursor, intermediate frataxin and/or mature frataxin, may be measured as a steady state level achieved after a period of treatment.
In one embodiment, the relative or absolute increase in level of frataxin is determined by comparing the total amount of frataxin, including frataxin precursor, intermediate frataxin and mature frataxin, present in the cell, subject, or patient before and after treatment in accordance with the invention. In another embodiment, the relative or absolute increase in level of frataxin is determined by comparing the amount of mature frataxin present in the cell, subject, or patient before and after treatment in accordance with the invention. In another embodiment, the relative or absolute increase in level of frataxin is determined by comparing the amount of frataxin precursor present in the cell, subject, or patient before and after treatment in accordance with the invention. In one embodiment, the relative or absolute increase in the level of frataxin is achieved by an increase in the expression of frataxin and is determined by comparing the amount of frataxin precursor present in the cell, subject or patient before and after treatment in accordance with the invention. In another embodiment, the relative or absolute increase in the level of frataxin is achieved by an increase in the expression of ,* PC72246
17 frataxin and is determined by comparing the amount of mature frataxin present in the cell, subject or patient before and after treatment in accordance with the invention.
Non-limiting examples of increases in the levels of frataxin that may be useful in certain embodiments disclosed herein are provided. For example, without wishing to be bound by theory, it is believed from observing patients or subjects with greater than from about 75 GAA triplet repeats to about 300 GAA triplet repeats, that a level of frataxin, including frataxin precursor, intermediate frataxin and / or mature frataxin, of about 20% of the level of frataxin, including frataxin precursor, intermediate frataxin and / or mature frataxin, of that seen in a healthy cell or subject results in late onset of the condition, disease or disorder, or the symptoms associated with the condition, disease or disorder. As such, it is expected that where a use of the present invention results in an increase in the level of frataxin, including frataxin precursor, intermediate frataxin and / or mature frataxin, to greater than about 20% of the level in a healthy cell or subject, the increase in frataxin, including frataxin precursor, intermediate frataxin and /
or mature frataxin, is expected to slow the advancement of the condition, disease or disorder or the symptoms associated with the condition, disease or disorder.
By way of further example, and again not wishing to be bound by theory, it is believed from observing heterozygous carriers that a level of about 50% of the level of frataxin, including frataxin precursor, intermediate frataxin and / or mature frataxin, of that seen in a healthy cell or subject results in asymptomatic state. As such, it is expected that where a use of the present invention results in an increase in the level of frataxin, including frataxin precursor, intermediate frataxin and / or mature frataxin, to greater than about 50% of the level in a healthy cell or subject, the increase in frataxin, including frataxin precursor, intermediate frataxin and / or mature frataxin, is expected to slow the advancement of the condition, disease or disorder or the symptoms associated with the conditions, disorder or disease, and potentially even prevent further advancement or lead to regression of the condition disorder or disease, or the symptoms associated with the condition, disease or disorder.
As used herein the term "healthy cell" is taken to mean a cell taken from a "healthy subject".
As used herein, the term "healthy subject" shall be taken to mean a subject who has not been diagnosed with a condition, disease or disorder associated with decreased = ' PC72246
18 level of frataxin or with decreased expression of frataxin. In one embodiment the healthy subject has been identified as not having Friedreich's ataxia. In another embodiment the healthy subject has been identified as not having late onset Friedreich's ataxia. In one embodiment, the healthy subject has been identified as having less than about 75 unstable homozygous GAA trinucleotide repeat expansion in the intron-1 on both of the alleles in the gene which encodes frataxin. In one embodiment, the healthy subject has been identified as having less than about unstable homozygous GAA trinucleotide repeat expansion in the intron-1 on both of the alleles in the gene which encodes frataxin. In one embodiment, the healthy subject has been identified as not having pathological mutations or repeat expansions on either allele of the intron-1 of the gene which encodes frataxin.
As used herein the term "decreased level of frataxin" shall be taken to mean that the level of frataxin, including frataxin precursor, intermediate frataxin and mature frataxin, present in a cell, subject or patient is lower than the level of frataxin, including frataxin precursor, intermediate frataxin and / or mature frataxin, observed in a healthy cell or subject. In one embodiment the decreased level of frataxin refers to a decreased level of frataxin precursor. In one embodiment the decreased level of frataxin refers to a decreased level of mature frataxin.
As used herein the term "decreased expression of frataxin" shall be taken to mean that the expression of frataxin, including frataxin precursor, intermediate frataxin and / or mature frataxin, in a cell, subject or patient is lower than the expression of frataxin, including frataxin precursor, intermediate frataxin and mature frataxin, observed in a healthy cell or subject. In one embodiment the decreased expression of frataxin refers to a decreased expression of frataxin precursor.
In one embodiment the cells associated with the methods and related uses of the present invention are those where a decrease in frataxin expression or a decrease in the level of frataxin in one or more of these cell types is thought to contribute specifically, in whole or in part, to the in vivo pathology of the patient or subject with the condition, disease or disorder associated with decreased frataxin expression or a decreased level of frataxin, or the associated symptoms. Such cells may include, but are not limited to, cerebellar neurons, neurons of the spinal tract, dorsal root ganglia and sensory neurons, aural and optic nerves, cardiomyocytes, and / or cells of the = PC72246
19 pancreas including islets. Without wishing to be bound by theory, cellular pathways believed to be modulated by changes in levels of frataxin include, but are not limited to, pathways that rely on functionality of an iron-sulphur protein such as the enzyme aconitase in the Kreb's/TCA cycle; succinate dehydrogenase subunit B (SDHB) in the electron transport chain; DNA polymerases (Pol a, 6 and e) and the major DNA
mutagenesis enzyme Pol important in DNA replication mechanisms and xanthine oxidase used in purine catabolism.
In one embodiment the cells associated with the methods of the invention, and related uses, are obtained from, or are present in, a subject diagnosed with a condition, disease or disorder associated with decreased expression of frataxin.
In one embodiment the cell is obtained from, or is present in, a subject diagnosed with a condition, disease or disorder associated with a decreased level of frataxin.
In one embodiment the cell is obtained from, or is present in, a subject diagnosed with Friedreich's ataxia. In one embodiment the cell is obtained from, or is present in, a human subject diagnosed with Friedreich's ataxia. In another embodiment the cell is obtained from, or is present in, a non-human species diagnosed with either a condition, disease or disorder associated with decreased expression of frataxin or condition, disease or disorder associated with a decreased level of frataxin or with Friedreich's ataxia, including genetic variants thereof. In one embodiment the cell is in vivo. In another embodiment the cell is ex vivo. In another embodiment the cell is in vitro.
As used herein the term "frataxin" shall be taken to mean one or more of the precursor polypeptide, intermediate frataxin and mature frataxin and the frataxin encoding nucleotide sequence and post translationally modified protein or polypeptide polymorphic variants, spliced variants, alleles, mutants and interspecies homologs that:
(1) have an amino acid sequence that has greater than about 90% amino acid sequence identity, for example 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98% or 99%
or greater amino acid sequence identity, preferably over a region of at least about 25, 50, 100, 200, 300, 400 or more amino acids or over the full-length to an amino acid sequence encoded by frataxin nucleic acid or to an amino acid sequence encoded by a frataxin polypeptide; (2) bind to antibodies eg polyclonal antibodies, raised against an immunogen comprising an amino acid sequence encoded by a frataxin nucleic acid (eg frataxin polynucleotides described herein), and conservatively modified variants thereof;

' PC72246 (3) specifically hybridise under stringent hybridization conditions to an anti-sense strand corresponding to a nucleic acid sequence that has greater than about 90% etc.
As used herein the term "gene which encodes frataxin" refers to a genomic region that encodes frataxin protein and / or controls the transcription of frataxin mRNA.
5 Thus, the term shall be taken to encompass coding sequences as well as any non-coding elements, including but not limited to, promoters, enhancers, silencers, introns and 5' and 3' untranslated regions. A gene which encodes frataxin may include flanking sequences 5' and / or 3' to a known annotated frataxin open reading frame, including, but not limited to, 1Kb, 2Kb, 3Kb, 4Kb, 5Kb, 6Kb, 7Kb, 8Kb, 9Kb, and / or 10Kb, or more 10 flanking the 5' and / or 3' end of a known annotated frataxin open reading frame. In one embodiment, a gene which encodes frataxin may be a human gene which encodes frataxin. In an alternative embodiment, a gene which encodes frataxin may be a corresponding homolog for a gene which encodes frataxin in a different species, including, but not limited to, for example a mouse.
15 Methods are available in the art for determining the level of frataxin, including frataxin precursor, intermediate frataxin and / or mature frataxin for example routine methods such as Western Blot analysis or commercially available dip stick testing kits such as those available from Abcam or Mitosciences .
Methods are available in the art for the identification of conditions, diseases or
20 disorders associated with a decreased expression of frataxin;
conditions, diseases or disorders associated with a decreased level of frataxin; for the identification of GAA
triplet repeat expansions on the intron-1 of the gene which encodes frataxin;
and / or which identify a point mutation or deletion on the gene which encodes frataxin including methods for the diagnosis of Friedreich's ataxia, for example the Friedreich's ataxia repeat expansion test available from Athena Diagnostics .
As used herein, the term "BET-family bromodomain" refers to members of the bromodomain family of proteins which contain two N-terminal bromodomains. This family includes bromodomain members known as BRD2, BRD3, BRD4 and BRDT.
These bromodomains are well known to share a common domain architecture featuring two amino-terminal bromodomains which exhibit high levels of sequence conservation, and a more divergent carboxy terminal recruitment domain.

' PC72246
21 As used herein, the term "BRD4" refers to BET-family bromodomain-containing protein 4, which is a member of the BET bromodomain family.
As used herein, the term "BET-family bromodomain inhibitor" shall be taken to mean a compound which shows inhibitory activity at one or more of the BET-family bromodomains, including by preventing the bromodomain from binding to acetyl-modified histone tails. In one embodiment the "BET-family bromodomain inhibitor" is a BRD4 inhibitor. In one embodiment the "BET-family bromodomain inhibitor" is a compound which inhibits bromodomain 1 of BRD4. In one embodiment the "BET-family bromodomain inhibitor" is a compound which inhibits bromodomain 2 of BRD4. In one embodiment the "BET-family bromodomain inhibitor" is a compound which inhibits both bromodomain 1 and bromodomain 2 of BRD4. Routine methods are available in the art to determine BET-family bromodomain activity such as the fluorescence binding assay disclosed herein and others.
As used herein, the term "IC50" refers to an amount, concentration or dosage of a particular test compound that achieves a 50% inhibition of a maximal response in an assay that measures such response. The value depends on the assay used.
BET-family bromodomain inhibitors The methods, uses, pharmaceutical usesand pharmaceutical compositions of the present invention utilize BET-family bromodomain inhibitors, or a pharmaceutically acceptable salt thereof.
Bromodomain-containing proteins are known to be of substantial biological interest, as components of transcription factor complexes and determinants of epigenetic memory. The BET-family (BRD2, BRD3, BRD4 and BRDT) shares a common domain architecture featuring two amino-terminal bromodomains that exhibit high levels of sequence conservation, and a more divergent carboxy-terminal recruitment domain (Filippakopoulos, P. et al., Nature 2010, 468, 1067-1073, incorporated by reference herein in its entirety for all purposes). BRD2 and BRD3 are reported to associate with histones along actively transcribed genes and may be involved in facilitating transcriptional elongation (Leroy et al, Mo/. Cell.
2008, 30, 51-60, incorporated by reference herein in its entirety for all purposes). It has also been reported that BRD4 or BRD3 may fuse with NUT (nuclear protein in testis) forming novel fusion oncogenes, BRD4-NUT or BRD3-NUT, in a highly malignant form of epithelial
22 neoplasia (French et al. Cancer Res., 2003, 63, 304-307 and French et al. J.
Clin.
Oncol. 2004, 22, 4135-4139, both of which are incorporated by reference herein in their entirety for all purposes). Data suggests that BRD-NUT fusion proteins contribute to carcinogensesis (French et al. Oncogene 2008, 27, 2237-2242, incorporated by reference herein in its entirety for all purposes). BRDT is believed to be uniquely expressed in the testes and ovary. All family members have been reported to have some function in controlling or executing aspects of the cell cycle, and have been shown to remain in complex with chromosomes during cell division ¨ suggesting a role in the maintenance of epigenetic memory. In addition some viruses make use of these proteins to tether their genomes to the host cell chromatin, as part of the process of viral replication (You et al. Cell 2004 117, 349-60, incorporated by reference herein in its entirety for all purposes). BRD4 appears to be involved in the recruitment of the pTEF-P complex to inducible genes, resulting in phosphorylation of RNA polymerase and increased transcriptional output (Hargreaves et al. Cell 2009 138, 129-145, incorporated by reference herein in its entirety for all purposes).
Bromodomain-containing protein 4 (BRD4) is a member of the BET-family that in yeast and animals contains two tandem bromodomains (BDI and BDII) and an extraterminal (ET) domain. BRD4 is a double bromodomain-containing protein that binds preferentially to acetylated chromatin. In humans, four BET proteins (BRD2, BRD3, BRD4 and BRDT) exhibit similar gene arrangements, domain organizations, and some functional properties (Wu, S. et al. J. Biol. Chem. 2007, 282, 13141-13145, incorporated by reference herein in its entirety for all purposes).
Small molecule BET-family bromodomain inhibitors have been described with a wide variety of different chemical scaffolds and different chemotype approaches are known to be useful (see for example Gamier et al. Expert Opinion Ther Patents 2014, 24(2), 185-199; Hewings et al. J Med Chem 2012, 55, 9393-413; and Owen et al.
Drug Disco Today: Technologies 2012, 10, 1016, all of which are incorporated by reference herein in their entirety for all purposes).
To date, small molecule BET-family bromodomain inhibitors have been mainly reported as being useful for treatment of inflammatory diseases and / or various cancers, but some have also been reported to be useful for the treatment of cardiovascular diseases and to treat male fertility. BET-family bromodomain inhibitors can be identified using routine methods available in the = `: PC72246
23 art such as the fluorescence polarization (FP) binding assay disclosed herein.
In one embodiment of the present invention, the BET-family bromodomain inhibitors may have an activity of about 55pM in the fluorescence polarization (FP) binding assay at either bromodomain 1, bromodomain 2 or both bromodomain 1 and 2.
In another embodiment, the BET-family bromodomain inhibitors useful in the present invention may have an activity of about 52pM in the fluorescence polarization (FP) binding assay disclosed herein at either bromodomain 1, bromodomain 2 or both bromodomain 1 and 2. In another embodiment, the BET-family bromodomain inhibitors useful in the present invention may have an activity of about 51pM in the fluorescence polarization (FP) binding assay disclosed herein at either bromodomain 1, bromodomain 2 or both bromodomain 1 and 2.
Examples of BET-family bromodomain inhibitors that may be used in the present invention include, but are not limited to compounds described below, including those which are in clinical development.
For example, small molecule BET-family bromodomain inhibitors with a diazepine chemical scaffold were described in International Patent application number PCT/EP2010/061518, published as W02011/054553 on 12th May 2011 which is incorporated by reference herein in its entirety for all purposes. This disclosure includes the compound I-BET762, and methods to prepare it:
N - N

N
41, /

0 ci I-BET762 is reported to be in clinical development for the treatment of various indications including acute & chronic lymphocytic leukemia; acute & chronic myelogenous leukemia; Hodgkin's & Non-Hodgkin's lymphoma; multiple myeloma;
myeloproliferative neoplasms; NUT mideline carcinoma; hematological carcinoma &
24 solid tumors; and atherosclerosis. Other disclosures relating to I-BET762 and analogues thereof include those described in International Patent application number PCT/EP2010/066697, published as W02011/054845 on 12th May 2011; International Patent application number PCT/EP2010/066696,published as W02011/054844 on 12th May 2011; International Patent application number PCT/EP2011/060179, published as W02011/161031 on 29th December 2011; and Nicodeme E et al. Nature, 2010, 468, 1119-23, all of which are incorporated by reference herein in their entirety for all purposes.
Small molecule BET-family bromodomain inhibitors with a thienodiazapine chemical scaffold were described in International Patent application number PCT/US2011/036701, published as W02011/143669 on 17th November 2011, which is incorporated by reference herein in its entirety for all purposes. This disclosure includes the compound JQ-1, which exists in two steroisomeric forms JQ-1(+) and JQ-1(-), and methods to prepare it:

S
Other disclosures relating to JQ-1 and analogues thereof include those described in International Patent application number PCT/US2011/036647, published as W02011/143651 on 17th November 2011; International Patent application number PCT/US2011/036667, published as W02011/143657 on 17th November 2011;
International Patent application number PCT/US2011/036672, published as W02011/143660 on 17th November 2011; and Filippakopoulos P et al. Nature, 2010, 468, 1067-73, all of which are incorporated by reference herein in their entirety for all purposes.

' PC72246 Other small molecule compounds with a thienodiazapine chemical scaffold were described in International Patent application number PCT/JP92/01198, published as WO 1994/06801 on 31st March 1994, which is incorporated by reference herein in its entirety for all purposes. This disclosure includes the racemic form of the compound 5 now known as CPI-203, also known as CPI-267203, and methods to prepare it:
N-N

S
CI
CPI-203 has since been reported to have BRD4 activity (Devaiah, B. N. et al.
Proc Nat!
Acad Sci USA 2012, 109(18), 6927-32, which is incorporated by reference herein in its entirety for all purposes) and is reported to be in preclinical development for a variety of 10 indications including cancer; hematological cancer; lymphoma; and Non-Hodgkin's lymphoma.
Yet further small molecule BET-family bromodomain inhibitors with thienodiazapine chemical scaffold were described in International Patent application number PCT/JP97/02817, published as W01998/011111 on 19th March 1998, which is 15 incorporated by reference herein in its entirety for all purposes. This disclosure includes the compound OTX-015, and methods to prepare it:

HO

S
OTX015 is reported to be in clinical development for various indications including leukemia; cancer; Crohn's disease; ulcerative colitis; hematological cancer;
acute lymphocytic leukemia; acute myelogenous leukemia; Non-Hodgkin's lymphoma;
multiple myeloma; and breast, lung, non-small cell, pancreas, prostate tumors. Further related compounds with this chemical scaffold are described in International Patent application PCT/JP2006/310709, published as W02006/129623 on 7th December 2006; and International Patent application PCT/JP2008/-73864, published as WO
2009/084693 on -th u July 2009, both of which are incorporated by reference herein in their entirety for all purposes.
Small molecule BET-family bromodomain inhibitors with a benzodiazepine chemical scaffold have been disclosed in International Patent application number PCT/EP2010/066696, published as W02011/054844 on 12th May 2011, which is incorporated by reference herein in its entirety for all purposes. This disclosure includes the compound GW-841819X, and methods to prepare it:
N N
çN 0 y /

=' PC72246 GW-841819X is reported to be in preclinical development for various indications including dyslipidemia; cancer; and hematological neoplasm.
Small molecule BET-family bromodomain inhibitors with a thienoazepine chemical scaffold have been described in International Patent application number PCT/US2011/063046, published as W02012/075383 on 7th June 2012, which is incorporated by reference herein in its entirety for all purposes. This disclosure includes the compound CP-0610, and methods to prepare it:
a CI
CPI-0610 is reported to be in clinical development for various indications including Hodgkin's & Non-Hodgkin's lymphoma; multiple myeloma; acute lymphocytic leukemia;
acute and chronic myelogenous leukemia; myelodysplastic syndrome; and myeloproliferative neoplasms.
Small molecule BET-family bromodomain inhibitors with an oxazolazepine chemical scaffold have been described in International Patent application number PCT/US2011/063046, published as W02012/075383 on 7th June 2012, which is incorporated by reference herein in its entirety for all purposes. This disclosure includes the compound CP1-232, and methods to prepare ' PC72246 N

=NH2 CI
Small molecule BET-family bromodomain inhibitors with a benzoazepine chemical scaffold have been described in International Patent application number PCT/EP2013/069902, published as W02014/048945 on 3rd April 2014, which is incorporated by reference herein in its entirety for all purposes. This disclosure includes the compound BET-BAY-002, and methods to prepare it:
N N
N = N
--il\s, \\_K
CI
BET-BAY-002 is reported to be in preclinical development for various indications including cancer.
Small molecule BET-family bromodomain inhibitors with a quinolone chemical scaffold have been described in International Patent application number PCT/EP2010/066695, published as W02011/054843 on 12th May 2011, which is incorporated by reference herein in its entirety for all purposes. This disclosure includes the compound I-BET-151, also known as GSK1210151, and methods to prepare it:

= -= PC72246 o N
NH

\o I-BET-151 is reported to be in preclinical development for various indications including leukemia; cancer; mixed lineage leukemia; hematological cancer; bone resorprtion; and inflammatory disease.
Small molecule BET-family bromodomain inhibitors with a quinazoline chemical scaffold have been described in International Patent application number PCT/CA2007/000146, published as W02008/092231 on 7th August 2008, which is incorporated by reference herein in its entirety for all purposes. This disclosure includes the compound RVX-208, and methods to prepare it:
OH

NH

RVX-208 is reported to be in clinical development for various indications including atherosclerosis; diabetes; acute coronary syndrome; myocardial Infarction;
Alzheimer's disease; stroke; mild cognitive impairment; low HDL cholesterol; and immunological diseases.
Small molecule BET-family bromodomain inhibitors with a benzopiperazine chemical scaffold have been described in International Patent application number PCT/EP2010/066695, published as W02011/054843 on 12th May 2011, which is ' PC72246 incorporated by reference herein in its entirety for all purposes. This disclosure includes the compound I-BET-726, also known as GSK1324726A, and methods to prepare it:
I- ....---- NFI
...---'--`,,--.,õ, --------N
0-*---;
I-BET-726 is reported to be in preclinical development for various indications including 5 cancer.
Small molecule BET-family bromodomain inhibitors with a thiophenpyran chemical scaffold have been described in International Patent application number PCT/US2009/031864, published as WO 2009/094560 on 30th July 2009, which is incorporated by reference herein in its entirety for all purposes. This disclosure includes 10 the compound SRX-2523, and methods to prepare it:

I
S
\ I 1 \

I

0 j SRX-2523 is reported to be in preclinical development for various indications including cancer and hematological neoplasm.

, PC72246 BET-family bromodomain inhibitors of the following formula:
AO
or pharmaceutically acceptable salts and solvates thereof, are disclosed in International Patent application PCT/IB2012/054211, published as WO 2013/027168 on 28th February 2013, which is incorporated by reference herein in its entirety for all purposes.
BET-family bromodomain inhibitors disclosed therein include N-(2-hydroxy-3-methylquinolin-6-yl)piperidine-1-sulfonamide HQN, HO
or a pharmaceutically acceptable salt or solvate thereof, which is specifically exemplified as example 46 in the above mentioned publication.
BET-family bromodomain inhibitors of the following formula (I):

' PC72246 Rx (I) or pharmaceutically acceptable salts thereof, are disclosed in International Patent application PCT/CN2011/002224, published as WO 2013/097052 on 4th July 2013;
and International Patent application PCT/CN2012/086357, published as WO

on 4th July 2013, both of which are incorporated by reference herein in their entirety for all purposes. BET-family bromodomain inhibitors disclosed therein include N44-(2,4-difluorophenoxy)-3-(6-methyl-7-oxo-6,7-dihydro-1H-pyrrolo[2, 3-c]pyrid in-yl)phenyliethanesulfonamide:

N

or a pharmaceutically acceptable salt thereof, which is specifically exemplified in the above mentioned publications. Further related publications include International Patent application PCT/US2014/044513, published as WO 2014/210425 on 31st December 2014, which is incorporated by reference herein in its entirety for all purposes.
Other BET-family bromodomain inhibitors which may also be used as disclosed herein include, but are not limited to, the following BET-family bromodomain inhibitors = " PC72246 which have been disclosed in the public domain as being in preclinical or clinical development BMS-986158 (Bristol Myers Squibb); RVX-297 (Resverlogix); RVS-2135 (Resverlogix); ZEN-3365 (Zenith); ZEN-3118 (Zenith); KM-601 (Kainos); SF-2535 (Signal Rx); AU-004 (Aurigene, Orion); ABBV-075 (Abbvie); TEN-010 (Tensha Therapeutics); and BAY1238097 (Bayer).
Yet further BET-family bromodomain inhibitors which may be used as described herein include, but are not limited to, compounds disclosed in the following international patent applications: triazolodiazepine derivatives disclosed in International Patent application PCT/US2012/036569, published as WO 2012/151512 on 8th November 2012; tetrahydroquinoline derivatives disclosed in International Patent application PCT/EP2010/066701, published as WO 2011/054848 on 12th May 2011;
imidazoquinoline derivatives disclosed in International patent application PCT/EP2010/066699, published as WO 2011/054846 on 12th May 2011; quinazolinone derivatives disclosed in International Patent application PCT/US2009/048457, published as WO 2009/158404 on 30th December 2009; quinazolinone derivatives disclosed in International Patent application PCT/US2010/031870, published as WO

on 28th October 2010; triazolopyridazine-6-amine derivatives disclosed in International Patent application PCT/U52012/042825, published as WO 2012/174487 on 20th December 2012; N-methylpyrrolopyridinones and N-methylpyrrolopyridazinone derivatives disclosed in International Patent application PCT/CN2012/086357, published as WO 2013/097601 on 4th July 2013; and bis aryl derivatives described in International Patent application PCT/US2010/026308, published as WO 2012/116170 on 30th August 2012, all of which are incorporated by reference herein in their entirety for all purposes.
Yet further BET ¨family bromodomain inhibitors which may be used in the present invention include those described in US provisional patent application serial number 62/181281, which was filed on 18th June 2015 which is incorporated by reference herein in its entirety for all purposes. This disclosure includes compounds of Formula I:

WN
Rlo Formula I
or a pharmaceutically acceptable salt thereof, wherein R1 is selected from the group consisting of:
(i) -C3-C7cycloalkyl optionally substituted with one, two, three or four E;
(ii) 4 to 7 membered heterocyclyl optionally substituted with one, two, three or four E, which said 4 to 7 membered heterocyclyl comprises one or two heteroatoms independently selected for each occurrence from the group consisting of N, 0 and S; and RiC
=
(iii) RiA is selected from the group consisting of (i) ¨C1-C6alkyl optionally substituted with one, two, three, four, five or six E;
(ii) -C3-C7cycloalkyl optionally substituted with one, two, three, four or five E;
(iii) phenyl optionally substituted with one, two, three, four or five E;
(iv) 4 to 7 membered heterocyclyl optionally substituted with one, two, three, four or five E, which said 4 to 7 membered heterocyclyl comprises one or two heteroatoms independently selected for each occurrence from the group consisting of N, 0 and S; and (v) 5 to 6 membered heteroaryl optionally substituted with one, two, three, four or five E, which said 5 to 6 membered heteroaryl comprises one, two or three heteroatoms independently selected for each occurrence from the group consisting of N, 0 and S;
R18 is selected from the group consisting of (i) ¨H; and 5 (ii) ¨Ci-C6alkyl optionally substituted with one, two, three, four, five or six E;
Ric is selected from the group consisting of (i) ¨H;
(ii) -CH3 optionally substituted with one, two, or three J;
10 (iii) ¨CH2CH3 optionally substituted with one, two, three, four or five J;
(iv) ¨CH2CH2CH3 optionally substituted with one, two, three, four, five, six or seven J; and (v) -CH(CH3)2 optionally substituted with one, two, three, four, 15 five, six or seven J;
R2A is selected from the group consisting of (i) ¨H;
(ii) ¨CH3 optionally substituted with one, two or three J;
(iii) ¨CH2CH3 optionally substituted with one, two, three, four, or five J;
20 and (iv) cyclopropyl optionally substituted with one, two, three, four or five J;
R2B is selected from the group consisting of (i) ¨C1-C6alkyl optionally substituted with one, two, three or four G;
(ii) ¨0C1-Cealkyl optionally substituted with one, two, three or four G;
25 (iii) ¨NH2, (iv) ¨NH(Ci-Csalkyl), which Ci-Csalkyl is optionally substituted with one, two, three or four G;
(v) ¨N(C1-C6alky1)2-, which C1-C6alkyl is, independently for each occurrence, opionally substituted with one, two, three or four G;
30 (vi) C3-05cycloalkyl optionally substituted with one, two, three or four G;
and (vii) 4 to 7 membered heterocyclyl optionally substituted with one, two, three or four G, which said 4 to 7 membered heterocyclyl comprises one or two heteroatoms independently selected for each occurrence from the group consisting of N, 0 and S;
W is selected from the group consisting of:

1¨N/ rµrn A4 (i) RaA
(ii) (iii) 1¨N\
(iv) RaB
(v) 5 ;and (vi) 4 to 7 membered heterocyclyl optionally substituted with one, two, three or four G, which said 4 to 7 membered heterocyclyl comprises one, two, three or four heteroatoms independently selected for each occurrence from the group consisting of N, 0 and S;
Y is selected from the group consisting of:
(i) ¨C H2¨ optionally substituted with one or two J;
(ii) ¨(CH2)2¨ optionally substituted with one, two, three or four J;
(iii) ¨(CH2)3¨ optionally substituted with one, two, three, four, five or six J; and (iv) ¨(CH2)4¨ optionally substituted with one, two, three, four, five, six, seven or eight J;
R3 is selected from the group consisting of:
(i) ¨H;
(ii) -CH3 optionally substituted with one, two, or three J;
(iii) ¨CH2CH3 optionally substituted with one, two, three, four or five J;

, = ' P072246 (iv) ¨CH2CH2CH3 optionally substituted with one, two, three, four, five, six or seven J; and (v) -CH(CH3)2 optionally substituted with one, two, three, four, five, six or seven J;
R4A is selected from the group consisting of (i) ¨H;
(ii) ¨01-C6alkyl optionally substituted with one, two, three or four G;
(iii) ¨CO2H;
(iv) ¨C(0)Ci-C6alkyl optionally substituted with one, two, three or four G;
(v) ¨C(0)0C1-C6alkyl optionally substituted with one, two, three or four G;
(vi) ¨C(0)NH2;
(vii) ¨C(0)NH(C1-C6alkyl) optionally substituted with one, two, three or four G;
(viii) ¨C(0)N(C1-C6alky1)2 optionally substituted with one, two, three or four G;
(ix) ¨C(0)NHSO2C1-C3alkyl optionally substituted with one, two, three or four G;
(x) ¨NH(C1-C3alkyl) optionally substituted with one, two, three or four G;
(xi) ¨N(C1-C3alky1)2 optionally substituted with one, two, three or four G;
(xii) ¨NHC(0)C1-C3alkyl optionally substituted with one, two, three or four G;
(xiii) ¨N(01-C3alkyl)C(0)C1-C3alkyl optionally substituted with one, two, three or four G;
(xiv) ¨NHSO2C1-C3alkyl optionally substituted with one, two, three or four G;
(xv) ¨N(C1-C3alkyl)S0201-C3alkyl optionally substituted with one, two, three or four G;
(xvi) ¨SO2NF12;

(xvii) ¨SO2NH(C1-C3alkyl) optionally substituted with one, two, three or four G;
(xviii) ¨SO2N(C1-C3alky1)2 optionally substituted with one, two, three or four G;
(xix) -C3-C7cycloalkyl optionally substituted with one, two, three or four G;
(xx) phenyl optionally substituted with one, two, three or four G;
(xxi) 4 to 7 membered heterocyclyl optionally substituted with one, two, three or four G, which said 4 to 7 membered heterocyclyl comprises one, two, three or four heteroatoms independently selected for each occurrence from the group consisting of N, 0 and S; and (xxii) 5 to 6 membered heteroaryl optionally substituted with one, two, three or four G, which said 5 to 6 membered heteroaryl ring comprises one, two, three or four heteroatoms independently selected for each occurrence from the group consisting of N, 0 and S;
R413 is selected from the group consisting of (i) ¨H;
(ii) ¨C1-C6alkyl optionally substituted with one, two, three or four G;
(iii) ¨C(0)C1-C6alkyl optionally substituted with one, two, three or four G;
(iv) ¨C(0)0C1-C6alkyl optionally substituted with one, two, three or four G;
(v) ¨C(0)NH2;
(vi) ¨C(0)NH(C1-C6alkyl) optionally substituted with one, two, three or four G;
(vii) ¨C(0)N(C1-C6alky1)2 optionally substituted with one, two, three or four G;
(viii) ¨C(0)NHSO2C1-C3alkyl optionally substituted with one, two, three or four G;
(ix) -C3-C7cycloalkyl optionally substituted with one, two, three or four G;
(x) phenyl optionally substituted with one, two, three or four G;

= PC72246 (xi) 4 to 7 membered heterocyclyl optionally substituted with one, two, three or four G, which said 4 to 7 membered heterocyclyl comprises one, two, three or four heteroatoms independently selected for each occurrence from the group consisting of N, 0 and S; and (xii) 5 to 6 membered heteroaryl optionally substituted with one, two, three or four G, which said 5 to 6 membered heteroaryl ring comprises one, two, three or four heteroatoms independently selected for each occurrence from the group consisting of N, 0 and S;
Fec is selected from the group consisting of (i) ¨H;
(ii) ¨C1-C6alkyl optionally substituted with one, two, three or four G;
(iii) -C3-C7cycloalkyl optionally substituted with one, two, three or four G;
(iv) phenyl optionally substituted with one, two, three or four G;
(v) 4 to 7 membered heterocyclyl optionally substituted with one, two, three or four G, which said 4 to 7 membered heterocyclyl comprises one, two, three or four heteroatoms independently selected for each occurrence from the group consisting of N, 0 and S; and (vi) 5 to 6 membered heteroaryl optionally substituted with one, two, three or four G, which said 5 to 6 membered heteroaryl ring comprises one, two, three or four heteroatoms independently selected for each occurrence from the group consisting of N, 0 and S;
R1 is independently selected for each occurrence from the group consisting of -H, -F, -Cl, -OH, -CN, -CH3, -CH2CH3, -CH2F, -CHF2, -CF3, -CF2CF3, -CH2OH, -OCH3, -OCH2F, -OCHF2, -0CF3, -SCH3, -SCH2F, -SCHF2, -SCF3 -N H2, -NH(CH3), and -N(CH3)2;
E is independently selected for each occurrence from the group consisting of:
(i) ¨OH;
(ii) ¨CN;
(iii) ¨CO2H;
(iv) ¨C(0)H;

(v) halo;
(vi) ¨C1-C3alkyl optionally substituted with one, two, three or four J;
(vii) -C1-C3alkylCO2H which ¨C1-C3alkyl is optionally substituted with one, two, three or four J;
5 (viii) ¨C3-C7cycloalkyl optionally substituted with one, two, three, four, five or six J;
(ix) ¨C1-C3alkyIC3-C6cycloalkyl optionally substituted with one, two, three, four, five or six J;
(x) ¨0C1-C3alkyl, optionally substituted with one, two, three or four J;
10 (xi) ¨0C3-C7cycloalkyl optionally substituted with one, two, three, four, five or six J;
(xii) ¨0C1-C3alkyIC3-C7cycloalkyl optionally substituted with one, two, three, four, five or six J;
(xiii) ¨SC1-C3alkyl, optionally substituted with one, two, three or four J;
15 (xiv) ¨SC3-C7cycloalkyl optionally substituted with one, two, three, four, five or six J;
(xv) ¨SC1-C3alkyIC3-C7cycloalkyl optionally substituted with one, two, three, four, five or six J;
(xvi) ¨C(0)C1-C3alkyl, optionally substituted with one, two, three or four J;
20 (xvii) ¨C(0)0C1-C3alkyl, optionally substituted with one, two, three or four J;
(xviii) ¨NH2;
(xix) ¨NH(C1-C3alkyl) optionally substituted with one, two, three or four J;
(xx) ¨N(Ci-C3alky1)2 which ¨C1-C3alkyl is, independently for each occurrence, optionally substituted with one, two, three or four J;
25 (xxi) ¨C(0)NH2;
(xxii) ¨C(0)NHC1-C3alkyl, optionally substituted with one, two, three or four J;
(xxiii) ¨C(0)N(C1-C3alky1)2, which ¨C1-C3alkyl is, independently for each occurrence, optionally substituted with one, two, three or four J;
(xxiv) ¨NHC(0)C1-C3alkyl, optionally substituted with one, two, three or four J;
30 (xxv) ¨S02(C1-C3alkyl), optionally substituted with one, two, three or four J;
(xxvi) ¨SO2NH(C1-C3alkyl), optionally substituted with one, two, three or four J;
(xxvii) ¨NHS02(C1-C3alkyl), optionally substituted with one, two, three or four J;
and (xxviii) phenyl optionally substituted with one, two, three, or four J;
G is independently selected for each occurrence from the group consisting of (i) ¨OH;
(ii) -CN;
(iii) ¨CO2H;
(iv) ¨C(0)H;
(v) halo;
(vi) ¨C1-C3alkyl, optionally substituted with one, two, three or four J;
(vii) ¨C1-C3alkylCO2H, which ¨C1-C3alkyl is optionally substituted with one, two, three or four J;
(viii) ¨C1-C3alkyIC3-C6cycloalkyl optionally substituted with one, two, three, four, five or six J;
(ix) ¨0C1-C3alkyl, optionally substituted with one, two, three or four J;
(x) ¨0C1-C3alkyIC3-C6cycloalkyl optionally substituted with one, two, three, four, five or six J;
(xi) ¨SC1-C3alkyl, optionally substituted with one, two, three or four J;
(xii) ¨SC1-C3alkyIC3-C6cycloalkyl optionally substituted with one, two, three, four, five or six J;
(xiii) ¨C(0)C1-C3alkyl, optionally substituted with one, two, three or four J;
(xiv) ¨C(0)0C1-C3alkyl, optionally substituted with one, two, three or four J;
(xv) (XVi) ¨NH(C1-C3alkyl), optionally substituted with one, two, three or four J;
(xvii) ¨N(C1-C3alky1)2, which ¨C1-C3alkyl is, independently for each occurrence, optionally substituted with one, two, three or four J;
(xviii) ¨C(0)NF12;
(xix) ¨C(0)NHC1-C3alkyl, optionally substituted with one, two, three or four J;
(xx) ¨C(0)N(C1-C3alky1)2, which ¨C1-C3alkyl is, independently for each occurrence, optionally substituted with one, two, three or four J;
(xxi) ¨NHC(0)C1-C3alkyl, optionally substituted with one, two, three or four J;
(xxii) ¨S02(C1-C3alkyl), optionally substituted with one, two, three or four J;
(xxiii) ¨SO2NH(Ci-C3alkyl), optionally substituted with one, two, three or four J;
and = PC72246 (xxiv) ¨NHS02(C1-C3alkyl) optionally substituted with one, two, three or four J;
and J is independently selected for each occurrence from the group consisting of -H, -F, -Cl, -OH, -CN, -CH3, -CH2CH3, -CH2F, -CHF2, -CF3, -CF2CF3, -CH2OH, -OCH3, -OCH2F, -OCHF2, -0CF3, -SCH3, -SCH2F, -SCHF2, -SCF3 -NH2, -NH(CH3), and -N(CH3)2.
A preferred Embodiment of the compounds of Formula I disclosed in US
provisional patent application serial number 62/181281 is also disclosed where:

= -,zz\R1B
RIC
=
R1 is RiA is selected from the group consisting of ¨CH2OCH3; phenyl; methoxyphenyl;
and pyridyl;
R1B is selected from the group consisting of methyl, ethyl, n-propyl, i-propyl, and ¨CH2OCH3;
Ric is _H;
R2A is selected from the group consisting of ¨CH3;
R2B is selected from the group consisting of methyl, ethyl, n-propyl, i-propyl, and -NH(CH3);

-1¨N/ R4A
W is Y is selected from the group consisting of -CH2- and -CH2CH2-;
R3 is ¨H;
R4A is selected from the group consisting of ¨CH3; ¨CO2H; and ¨C(0)NHCH3;
and R1 is ¨H, or a pharmaceutically acceptable salt thereof.

Specific examples of BET-family bromodomain inhibitors, including their methods of preparation, described in US provisional patent application serial number 62/181281, which was filed on 18th June 2015, include:
N-{6-[acetyl(methypamino]-4-[(1S)-1-(2-methoxyphenypethyl]-3-oxo-3,4-dihydropyrido[2,3-b]pyrazin-2-y1}-beta-alanine;
N-{64acetyl(ethyl)amino]-4-[(1S)-1-(2-methoxyphenypethyl]-3-oxo-3,4-dihydropyrido[2,3-b]pyrazin-2-y1}-beta-alanine;
N-{6-[acetyl(methyl)amino]-3-oxo-4-[(1S)-1-phenylpropyl]-3,4-dihydropyrido[2,3-b]pyrazin-2-y1}-beta-alanine;
N-{6-[(hydroxyacetyl)(methyl)amino]-3-oxo-4-[(1S)-1-phenylpropyl]-3,4-dihydropyrido[2,3-b]pyrazin-2-yI}-beta-alanine;
N-{4-[(1S)-1-(2-methoxyphenypethy1]-2-(methylamino)-3-oxo-3,4-dihydropyrido[2,3-b]pyrazin-6-y1}-N-methylacetamide;
N-{6-[methyl(2-methylpropanoyDamino]-3-oxo-4-[(1S)-1-phenylpropyl]-3,4-dihydropyrido[2,3-b]pyrazin-2-y1}-beta-alanine;
N-{6-[butanoyl(methyl)amino]-3-oxo-4-[(1S)-1-phenylpropy1]-3,4-dihydropyrido[2,3-b]pyrazin-2-y1}-beta-alanine;
N-{6-[(cyclobutylcarbonyl)(methyl)amino]-3-oxo-4-[(1S)-1-phenylpropyl]-3,4-dihydropyrido[2,3-b]pyrazin-2-yll-beta-alanine;
N-{64methyl(methylcarbamoyl)amino]-3-oxo-4-[(1S)-1-phenylpropyl]-3,4-dihydropyrido[2,3-b]pyrazin-2-y1}-beta-alanine;
N-{6-[methyl(propanoyl)amino]-3-oxo-4-[(1S)-1-phenylpropy1]-3,4-dihydropyrido[2,3-b]pyrazin-2-yll-beta-alanine;
N-{6-[methyl(propanoyDamino]-3-oxo-4-[(1S)-1-(pyridin-2-yl)propyl]-3,4-dihydropyrido[2,3-b]pyrazin-2-yI}-beta-alanine;
N-{64methyl(propanoyDaminol-3-oxo-4-(1-phenylcyclobutyl)-3,4-dihydropyrido[2,3-b]pyrazin-2-y1}-beta-alanine;
N-{4-(2,5-diethylcyclopentyI)-6-[methyl(propanoyl)amino]-3-oxo-3,4-dihydropyrido[2,3-b]pyrazin-2-y1}-beta-alanine;
N-{64acetyl(methyl)amino]-4-[(1R)-2-methoxy-1-phenylethy1]-3-oxo-3,4-dihydropyrido[2,3-b]pyrazin-2-y1}-beta-alanine;
N-{6-[methyl(propanoyl)amino]-3-oxo-4-[(3S,4S)-4-phenyltetrahydrofuran-3-y1]-3,4-d ihyd ropyrido[2,3-b]pyrazin-2-yll-beta-ala nine;

= ,N PC72246 N-{6-[acetyl(methypamino]-4-[(2R)-1-methoxybutan-2-y1]-3-oxo-3,4-dihydropyrido[2,3-b]pyrazin-2-y1}-beta-alanine;
N-{4-[(2R)-1-methoxypentan-2-y1]-6-[methyl(propanoyl)amino]-3-oxo-314-dihydropyrido[2,3-b]pyrazin-2-y1}-beta-alanine;
N-(4-[(2R)-1-methoxybutan-2-y1]-2-{[2-(methylamino)-2-oxoethyl]amino}-3-oxo-3,4-dihydropyrido[2,3-b]pyrazin-6-y1)-N-methylpropanamide;
N44-(1,3-dimethoxypropan-2-y1)-2-{[2-(rnethylamino)-2-oxoethyl]amino}-3-oxo-3, d ihyd ropyrido[2,3-b]pyrazin-6-y1]-N-methylpropanamide;
N-[4-(1 , 3-d imethoxypropan-2-y1)-2-(methylamino)-3-oxo-3,4-dihyd ropyrido[2 ,3-b]pyrazin-6-y1]-N-methylpropanamide;
N-{2-(acetylamino)-3-oxo-4-[(1S)-1-phenylpropy1]-3,4-dihydropyrido[2,3-b]pyrazin-6-y1}-N-methylacetamide;
N-{6-Rdimethylcarbamoy1)(methypamino]-3-oxo-4-[(1S)-1-phenylpropyl]-3,4-dihydropyrido[2,3-13]pyrazin-2-y1}-beta-alanine;
N-{6-[methyl(propanoyl)amino]-4-[(1S)-2-methyl-1-(pyridin-2-yl)propyl]-3-oxo-3,4-dihydropyrido[2,3-b]pyrazin-2-y1}-beta-alanine;
N-{4-(1-cyclopentylcyclopropy1)-64methyl(propanoyDamino]-3-oxo-3 14-di hyd ropyrido[2 ,3-b]pyrazin-2-y1}-beta-alanine;
N-(6-{[(3, 3-d imethylcyclobutypcarbonyl](methypam ino}-3-oxo-4-[(1S)-1-phenylpropyl]-3,4-dihydropyrido[2,3-b]pyrazin-2-y1)-beta-alanine;
N-(6-{[(3, 3-d ifl uorocyclobutyl)carbonyl](methyDam ino}-3-oxo-4-[(1S)-1-phenylp ropy1]-3,4-dihyd ropyrido[2,3-b]pyrazin-2-y1)-beta-alanine;
N-{6-[methyl(oxetan-3-ylcarbonypamino]-3-oxo-4-[(1S)-1-phenylpropy1]-3,4-dihydropyrido[2,3-b]pyrazin-2-y1}-beta-alanine;
N-methyl-N-(24[3-(methylamino)-3-oxopropyl]amino}-3-oxo-4-[(1S)-1-phenylpropyl]-3,4-dihydropyrido[2,3-b]pyrazin-6-y1)oxetane-2-carboxamide;
N-methyl-N-(2-1[2-(methylamino)-2-oxoethyl]amino}-3-oxo-4-[(1S)-1-phenylpropy1]-3,4-dihydropyrido[2,3-b]pyrazin-6-yppropanamide;
N-methyl-N-(24[2-(methylamino)-2-oxoethyl]amino}-4-[(1S)-2-methyl-1-(pyridin-2-yl)propy1]-3-oxo-3,4-dihydropyrido[2,3-b]pyrazin-6-yl)propanamide;
N3-{4-[(1R)-2-methoxy-1-phenylethyl]-6-[methyl(propanoyDamino]-3-oxo-3,4-dihydropyrido[2,3-b]pyrazin-2-y1}-N-methyl-beta-alaninamide;

N-methyl-N3-{6-[methyl(propanoyDamino]-3-oxo-4-[(1S)-1-phenylpropyl]-3,4-dihydropyrido[2,3-b]pyrazin-2-y1}-beta-alaninamide;
N-methyl-N3-{64methyl(propanoyl)amino]-3-oxo-4-[(1S)-1-(pyridin-2-yl)propyl]-3,4-dihydropyrido[2,3-b]pyrazin-2-y1}-beta-alaninamide; and 5 N-methyl-N3-{64methyl(propanoyl)amino]-4-[(1S)-2-methyl-1-(pyridin-2-yppropyl]-3-oxo-3,4-dihydropyrido[2,3-b]pyrazin-2-y1}-beta-alaninamide or a pharmaceutically acceptable salt thereof.
Preferred BET-family bromodomain inhibitors described in US provisional patent 10 application serial number 62/181281, which was filed on 18th June 2015, include:
N-{6-[methyl(propanoyl)amino]-3-oxo-4-[(1S)-1-phenylpropy1]-3,4-dihydropyrido[2,3-b]pyrazin-2-y1}-beta-alanine;
N-methyl-N3-{64methyl(propanoyDarnino]-3-oxo-4-[(1S)-1-phenylpropyl]-3,4-dihydropyrido[2,3-b]pyrazin-2-y1}-beta-alaninamide;
15 N-methyl-N-(24[2-(methylamino)-2-oxoethyllamino}-3-oxo-4-[(1S)-1-phenylpropyll-3,4-dihydropyrido[2,3-b]pyrazin-6-yppropanamide;
N3-{4-[(1R)-2-methoxy-1-phenylethyl]-6-[methyl(propanoyl)amino]-3-oxo-3,4-dihydropyrido[2,3-b]pyrazin-2-y1}-N-methyl-beta-alaninamide;
N-methyl-N-(24[3-(methylamino)-3-oxopropyl]amino}-3-oxo-4-[(1S)-1-phenylpropyl]-3,4-20 dihydropyrido[2,3-b]pyrazin-6-yl)oxetane-2-carboxamide;
N-{6-[methyl(methylcarbamoyl)amino]-3-oxo-4-[(1S)-1-phenylpropy1]-3,4-dihydropyrido[2,3-b]pyrazin-2-y1}-beta-alanine; and N-methyl-N3-{64methyl(propanoypamino]-4-[(1S)-2-methyl-1-(pyridin-2-y1)propyl]-3-oxo-3,4-dihydropyrido[2,3-b]pyrazin-2-y1}-beta-alaninamide 25 or a pharmaceutically acceptable salt thereof.
An especially preferred BET-family bromodomain inhibitor described in US
provisional patent application serial number 62/181281, which was filed on 18th June 2015, is:
N-{6-[methyl(propanoyl)amino]-3-oxo-4-[(1S)-1-phenylpropy1]-3,4-dihydropyrido[2,3-b]pyrazin-2-y1}-beta-alanine 30 or a pharmaceutically acceptable salt thereof.

Still further BET ¨family bromodomain inhibitors which may be used in the present invention include those described in US provisional patent application serial number 62/181281, which was filed on 18th June 2015 the disclosure of which is hereby incorporated by reference in its entirety for all purposes. This disclosure includes compounds of Formula II:

WN
Formula ll or a pharmaceutically acceptable salt thereof, wherein R1 is selected from the group consisting of:
(i) -C3-C7cycloalkyl optionally substituted with one, two, three or four E;
(ii) 4 to 7 membered heterocyclyl optionally substituted with one, two, three or four E, which said 4 to 7 membered heterocyclyl comprises one or two heteroatoms independently selected for each occurrence from the group consisting of N, 0 and S; and plA
_2zzc,R1B
WC
(iii) =
R1A is selected from the group consisting of (i) ¨C1-C6alkyl optionally substituted with one, two, three, four, five or six E;
(ii) -C3-C7cycloalkyl optionally substituted with one, two, three, four or five E;
(iii) phenyl optionally substituted with one, two, three, four or five E;
(iv) 4 to 7 membered heterocyclyl optionally substituted with one, two, three, four or five E, which said 4 to 7 membered heterocyclyl comprises one or two heteroatoms independently selected for each occurrence from the group consisting of N, 0 and S; and (v) 5 to 6 membered heteroaryl optionally substituted with one, two, three, four or five E, which said 5 to 6 membered heteroaryl comprises one, two or three heteroatoms independently selected for each occurrence from the group consisting of N, 0 and S;
R1B is selected from the group consisting of (i) ¨H; and (ii) ¨C1-C6alkyl optionally substituted with one, two, three, four, five or six E;
Ric is selected from the group consisting of (i) ¨H;
(ii) -CH3 optionally substituted with one, two, or three J;
(iii) ¨CH2CH3 optionally substituted with one, two, three, four or five J;
(iv) ¨CH2CH2CH3 optionally substituted with one, two, three, four, five, six or seven J; and (v) -CH(CH3)2 optionally substituted with one, two, three, four, five, six or seven J;
W is selected from the group consisting of:

¨N

(ii) (iii) 1¨N( (iv) R4B

' PC72246 (v) ;and (vi) 4 to 7 membered heterocyclyl optionally substituted with one, two, three or four G, which said 4 to 7 membered heterocyclyl comprises one, two, three or four heteroatoms independently selected for each occurrence from the group consisting of N, 0 and S;
Y is selected from the group consisting of:
(i) ¨C H2¨ optionally substituted with one or two J;
(ii) ¨(CH2)2¨ optionally substituted with one, two, three or four J;
(iii) ¨(CH2)3¨ optionally substituted with one, two, three, four, five or six J; and (iv) ¨(CH2)4¨ optionally substituted with one, two, three, four, five, six, seven or eight J;
R3 is selected from the group consisting of:
(i) ¨H;
(ii) -CH3 optionally substituted with one, two, or three J;
(iii) ¨CH2CH3 optionally substituted with one, two, three, four or five J;
(iv) ¨CH2CH2CH3 optionally substituted with one, two, three, four, five, six or seven J; and (v) CH(CH3)2 optionally substituted with one, two, three, four, five, six or seven J;
134A is selected from the group consisting of (i) ¨H;
(ii) ¨C1-C6alkyl optionally substituted with one, two, three or four G;
(iii) ¨CO2H;
(iv) ¨C(0)C1-C6alkyl optionally substituted with one, two, three or four G;
(v) ¨C(0)0C1-C6alkyl optionally substituted with one, two, three or four G;
(vi) ¨C(0)NH2;
(vii) ¨C(0)NH(C1-C6alkyl) optionally substituted with one, two, three or four G;

= õ === PC72246 (viii) ¨C(0)N(Ci-C6alky1)2 optionally substituted with one, two, three or four G;
(ix) ¨C(0)NHSO2C1-C3alkyl optionally substituted with one, two, three or four G;
(x) ¨NH(C1-C3alkyl) optionally substituted with one, two, three or four G;
(xi) ¨N(C1-C3alky1)2 optionally substituted with one, two, three or four G;
(xii) ¨NHC(0)C1-C3alkyl optionally substituted with one, two, three or four G;
(xiii) ¨N(C1-C3alkyl)C(0)C1-C3alkyl optionally substituted with one, two, three or four G;
(xiv) ¨NHSO2C1-C3alkyl optionally substituted with one, two, three or four G;
(xv) ¨N(C1-C3alkyl)S02C1-C3alkyl optionally substituted with one, two, three or four G;
(xvi) ¨SO2NH2;
(xvii) ¨SO2NH(C1-C3alkyl) optionally substituted with one, two, three or four G;
(xviii) ¨SO2N(C1-C3alky1)2 optionally substituted with one, two, three or four G;
(xix) -C3-C7cycloalkyl optionally substituted with one, two, three or four G;
(xx) phenyl optionally substituted with one, two, three or four G;
(xxi) 4 to 7 membered heterocyclyl optionally substituted with one, two, three or four G, which said 4 to 7 membered heterocyclyl comprises one, two, three or four heteroatoms independently selected for each occurrence from the group consisting of N, 0 and S; and (xxii) 5 to 6 membered heteroaryl optionally substituted with one, two, three or four G, which said 5 to 6 membered heteroaryl ring comprises one, two, three or four heteroatoms independently selected for each occurrence from the group consisting of N, 0 and S;
R4B is selected from the group consisting of (i) ¨H;
(ii) ¨C1-C6alkyl optionally substituted with one, two, three or four G;
(iii) ¨C(0)C1-C6alkyl optionally substituted with one, two, three or four G;
5 (iv) ¨C(0)0C1-C6alkyl optionally substituted with one, two, three or four G;
(v) ¨C(0)NF12;
(vi) ¨C(0)NH(C1-C6alkyl) optionally substituted with one, two, three or four G;
10 (vii) ¨C(0)N(C1-C6alky1)2 optionally substituted with one, two, three or four G;
(viii) ¨C(0)NHSO2C1-C3alkyl optionally substituted with one, two, three or four G;
(ix) -C3-C7cycloalkyl optionally substituted with one, two, three or four 15 G;
(x) phenyl optionally substituted with one, two, three or four G;
(xi) 4 to 7 membered heterocyclyl optionally substituted with one, two, three or four G, which said 4 to 7 membered heterocyclyl comprises one, two, three or four heteroatoms independently selected for 20 each occurrence from the group consisting of N, 0 and S; and (xii) 5 to 6 membered heteroaryl optionally substituted with one, two, three or four G, which said 5 to 6 membered heteroaryl ring comprises one, two, three or four heteroatoms independently selected for each occurrence from the group consisting of N, 0 and 25 S;
R4c is selected from the group consisting of (i) ¨H;
(ii) ¨C1-C6alkyl optionally substituted with one, two, three or four G;
(iii) -C3-C7cycloalkyl optionally substituted with one, two, three or four 30 G;
(iv) phenyl optionally substituted with one, two, three or four G;
(v) 4 to 7 membered heterocyclyl optionally substituted with one, two, three or four G, which said 4 to 7 membered heterocyclyl comprises .= PC72246 one, two, three or four heteroatoms independently selected for each occurrence from the group consisting of N, 0 and S; and (vi) 5 to 6 membered heteroaryl optionally substituted with one, two, three or four G, which said 5 to 6 membered heteroaryl ring comprises one, two, three or four heteroatoms independently selected for each occurrence from the group consisting of N, 0 and S;
R1 is independently selected for each occurrence from the group consisting of -H, -F, -Cl, -OH, -CN, -CH3, -CH2CH3, -CH2F, -CHF2, -CF3, -CF2CF3, -CH2OH, -OCH3, -OCH2F, -OCHF2, -0CF3, -SCH3, -SCH2F, -SCHF2, -SCF3 -NH2, -NH(CH3), and -N(CH3)2, E is independently selected for each occurrence from the group consisting of:
(i) ¨OH;
(ii) ¨CN;
(iii) ¨CO2H;
(iv) ¨O(0)H;
(v) halo;
(vi) ¨C1-C3alkyl optionally substituted with one, two, three or four J;
(vii) -01-C3alkylCO2H which ¨C1-C3alkyl is optionally substituted with one, two, three or four J;
(viii) ¨03-C7cycloalkyl optionally substituted with one, two, three, four, five or six J;
(ix) ¨01-C3alky1C3-C6cycloalkyl optionally substituted with one, two, three, four, five or six J;
(x) ¨001-C3alkyl, optionally substituted with one, two, three or four J;
(xi) ¨0C3-C7cycloalkyl optionally substituted with one, two, three, four, five or six J;
(xii) ¨0C1-C3alkyIC3-C7cycloalkyl optionally substituted with one, two, three, four, five or six J;
(xiii) ¨S01-C3alkyl, optionally substituted with one, two, three or four J;
(xiv) ¨S03-C7cycloalkyl optionally substituted with one, two, three, four, five or six J;

= PC72246 (xv) ¨SC1-C3alkyIC3-C7cycloalkyl optionally substituted with one, two, three, four, five or six J;
(xvi) ¨C(0)Ci-C3alkyl, optionally substituted with one, two, three or four J;
(xvii) ¨C(0)0C1-C3alkyl, optionally substituted with one, two, three or four J;
(xviii) ¨NH2;
(xix) ¨NH(C1-C3alkyl) optionally substituted with one, two, three or four J;
(xx) ¨N(C1-O3alky1)2 which ¨C1-C3alkyl is, independently for each occurrence, optionally substituted with one, two, three or four J;
(xxi) ¨C(0)NH2;
(xxii) ¨C(0)NHCi-C3alkyl, optionally substituted with one, two, three or four J;
(xxiii) ¨C(0)N(C1-C3alky1)2, which ¨C1-C3alkyl is, independently for each occurrence, optionally substituted with one, two, three or four J;
(xxiv) ¨NHO(0)C1-C3alkyl, optionally substituted with one, two, three or four J;
(xxv) ¨S02(C1-C3alkyl), optionally substituted with one, two, three or four J;
(xxvi) ¨SO2NH(C1-C3alkyl), optionally substituted with one, two, three or four J;
(xxvii) ¨NHS02(C1-C3alkyl), optionally substituted with one, two, three or four J;
and (xxviii) phenyl optionally substituted with one, two, three, or four J;
G is independently selected for each occurrence from the group consisting of (i) ¨OH
(ii) -ON;
(iii) ¨CO2H;
(iv) ¨O(0)H;
(v) halo;
(vi) ¨01-C3alkyl, optionally substituted with one, two, three or four J;¨

(vii) ¨C1-C3alkylCO2H, which ¨C1-C3alkyl is optionally substituted with one, two, three or four J;
(viii) ¨01-C3alky1C3-C6cycloalkyl optionally substituted with one, two, three, four, five or six J;
(ix) ¨001-C3alkyl, optionally substituted with one, two, three or four J;
(x) ¨001-O3alky1C3-C6cycloalkyl optionally substituted with one, two, three, four, five or six J;

= PC72246 (xi) ¨SC1-C3alkyl, optionally substituted with one, two, three or four J;
(xii) ¨SC1-C3alkyIC3-C6cycloalkyl optionally substituted with one, two, three, four, five or six J;
(xiii) ¨C(0)C1-C3alkyl, optionally substituted with one, two, three or four J;
(xiv) ¨C(0)0C1-C3alkyl, optionally substituted with one, two, three or four J;
(xv) ¨NH2;
(xvi) ¨NH(C1-C3alkyl), optionally substituted with one, two, three or four J;
(xvii) ¨N(C1-C3alky1)2, which ¨C1-C3alkyl is, independently for each occurrence, optionally substituted with one, two, three or four J;
(xviii) ¨C(0)NH2;
(xix) ¨C(0)NHC1-C3alkyl, optionally substituted with one, two, three or four J;
(xx) ¨C(0)N(C1-C3alky1)2, which ¨C1-C3alkyl is, independently for each occurrence, optionally substituted with one, two, three or four J;
(xxi) ¨NHC(0)Ci-C3alkyl, optionally substituted with one, two, three or four J;
(xxii) ¨S02(C1-C3alkyl), optionally substituted with one, two, three or four J;
(xxiii) ¨SO2NH(C1-C3alkyl), optionally substituted with one, two, three or four J;
and (xxiv) ¨NHS02(C1-C3alkyl) optionally substituted with one, two, three or four J;
and J is independently selected for each occurrence from the group consisting of -H, -F, -CI, -OH, -CN, -CH3, -CH2CH3, -CH2F, -CHF2, -CF3, -CF2CF3, -CH2OH, -OCH3, -OCH2F, -OCHF2, -0CF3, -SCH3, -SCH2F, -SCHF2, -SCF3 -NH2, -NH(CH3), and -N(CH3)2.
A preferred Embodiment of these compounds of Formula II is disclosed in US
provisional patent application serial number 62/181281, where:
R1A lB
Ric =
R1 is RiA is selected from the group consisting of ¨CH2OCH3; phenyl; methoxyphenyl;
and pyridyl;

R1B is selected from the group consisting of methyl, ethyl, n-propyl, i-propyl, and ¨CH2OCH3;
Ric is ¨H;

W is Y=
Y is selected from the group consisting of -CH2- and -CH2CH2-;
IR3 is ¨H;
IR"' is selected from the group consisting of ¨CH3; ¨CO2H; and ¨C(0)NHCH3;
and R1 is ¨H, or a pharmaceutically acceptable salt thereof.
Specific examples of BET-family bromodomain inhibitors, including their methods of preparation, of Formula II described in US provisional patent application serial number 62/181281, which was filed on 18th June 2015 include:
6-(3,5-dimethy1-1,2-oxazol-4-y1)-4-[(1S)-1-(2-methoxyphenypethyl]-2-{[2-(morpholin-4-ypethyl]aminolpyrido[2,3-b]pyrazin-3(4H)-one;
6-(3,5-dimethy1-1,2-oxazol-4-y1)-4-(2-ethoxybenzy1)-2-{[2-(morpholin-4-ypethyl]amino}pyrido[2,3-b]pyrazin-3(4H)-one;
4-benzy1-6-(3,5-dimethy1-1,2-oxazol-4-y1)-2-{[2-(morpholin-4-ypethyl]amino}pyrido[2,3-b]pyrazin-3(4H)-one;
6-(3,5-dimethy1-1,2-oxazol-4-y1)-2-{[2-(morpholin-4-ypethyl]amino}-4-[(1R)-1-phenylpropyl]pyrido[2,3-b]pyrazin-3(4H)-one;
6-(3,5-dimethy1-1,2-oxazol-4-y1)-2-{[2-(morpholin-4-ypethyl]aminol-4-[(1S)-1-phenylpropyl]pyrido[2,3-b]pyrazin-3(4H)-one;
6-(3,5-dimethy1-1,2-oxazol-4-y1)-2-{[2-(morpholin-4-ypethyl]amino}-4-[(1S)-1-phenylethyl]pyrido[2,3-b]pyrazin-3(4H)-one;
N-{6-(3,5-dimethy1-1,2-oxazol-4-y1)-3-oxo-4-[(1S)-1-phenylethyl]-3,4-dihydropyrido[2,3-b]pyrazin-2-yllglycine;
N-{6-(3,5-dimethy1-1,2-oxazol-4-y1)-3-oxo-4-[(1S)-1-phenylethyl]-3,4-dihydropyrido[2,3-b]pyrazin-2-yll-beta-alanine;

N-{6-(3,5-dimethy1-1,2-oxazol-4-y1)-4-[(1S)-1-(2-methoxyphenypethyl]-3-oxo-3,4-dihydropyrido[2,3-b]pyrazin-2-y1}-beta-alanine;
N-{6-(3,5-dimethy1-1,2-oxazol-4-y1)-3-oxo-4-[(1S)-1-phenylpropyl]-3,4-dihydropyrido[2,3-b]pyrazin-2-yll-beta-alanine;
5 N-{6-(3, 5-d imethy1-1,2-oxazol-4-y1)-3-oxo-4-[(1S)-1-(pyrimidin-2-y0propylj-3,4-dihydropyrido[2,3-b]pyrazin-2-y1}-beta-alanine;
N-{6-(3,5-dimethy1-1,2-oxazol-4-y1)-4-[(2S)-1-methoxybutan-2-y1]-3-oxo-3,4-dihydropyrido[2,3-b)pyrazin-2-y1}-beta-alanine;
,4-N-[4-(1,3-dimethoxypropan-2-y1)-6-(3,5-dimethy1-1,2-oxazol-4-y1)-3-oxo-3,4-dihydropyrido[2,3-b]pyrazin-2-y1]-beta-alanine;
N46-(3,5-dimethy1-1,2-oxazol-4-y1)-3-oxo-4-(tetrahydro-2H-pyran-4-y1)-3,4-dihydropyrido[2,3-13]pyrazin-2-y1]-beta-alanine;
N3-{6-(3,5-dimethy1-1,2-oxazol-4-y1)-3-oxo-4-[(1S)-1-phenylethyl]-3,4-dihydropyrido[2,3-13]pyrazin-2-y1}-N-(methylsulfony1)-beta-alaninamide;
6-(3,5-dimethy1-1,2-oxazol-4-y1)-4-[(1S)-1-phenylethyl]-2-{[2-(1H-tetrazol-5-ypethyl]amino}pyrido[2,3-13]pyrazin-3(4H)-one;
N-{6-(3,5-dimethy1-1,2-oxazol-4-y1)-3-oxo-4-[(1S)-1-phenylbutyl]-3,4-dihydropyrido[2,3-b]pyrazin-2-yll-beta-alanine;
N-{6-(3,5-dimethy1-1,2-oxazol-4-y1)-4-[(1S)-2-methyl-1-phenylpropyl]-3-oxo-3,4-dihyd ropyrido[2,3-b]pyrazin-2-yll-beta-alanine;
N-{6-(3,5-dimethy1-1,2-oxazol-4-y1)-4-[(1S)-2-methyl-1-(pyridin-2-y1)propyl]-3-oxo-3,4-dihydropyrido[2,3-b]pyrazin-2-y1}-beta-alanine;
N-{4-[(1S)-1-cyclohexylethyl]-6-(3, 5-d imethy1-1,2-oxazol-4-y1)-3-oxo-3,4-dihydropyrido[2,3-b]pyrazin-2-y1}-beta-alanine;
N46-(3, 5-d imethy1-1,2-oxazol-4-y1)-3-oxo-4-(pentan-3-y1)-3 ,4-d I hydropyrid o[2,3-b]pyrazin-2-y11-beta-alanine; and N2-{6-(3,5-d imethy1-1,2-oxazol-4-y1)-4-[(1S)-1-(2-methoxyphenypethyl]-3-oxo-3,4-dihydropyrido[2,3-b]pyrazin-2-yll-N-methylglycinamide or a pharmaceutically acceptable salt thereof.

= PC72246 A preferred BET-family bromodomain inhibitor of Formula II described in US
provisional patent application serial number 62/181281, which was filed on 18th June 2015, is:
N-{6-(3,5-d imethy1-1,2-oxazol-4-y1)-4-[(2R)-1-methoxybutan-2-y1]-3-oxo-3,4-dihydropyrido[2,3-b]pyrazin-2-y1}-beta-alanine or a pharmaceutically acceptable salt thereof.
As used herein, unless otherwise noted, "alkyl" whether used alone or as part of a substituent group refers to a saturated straight or branched hydrocarbon chain (ie a substituent obtained from a hydrocarbon by removal of a hydrogen) having from one to twenty carbon atoms or any number within this range, for example, from one to six carbon atoms, from one to four carbon atoms or from one to three carbon atoms.

Designated numbers of carbon atoms (e.g. C1.6) shall refer independently to the number of carbon atoms in an alkyl moiety or to the alkyl portion of a larger alkyl-containing substituent. Examples of alkyl groups include, but are not limited to, methyl, ethyl, n-propyl, iso-propyl, n-butyl, sec-butyl, iso-butyl, tert-butyl, pentyl, isoamyl, hexyl and the like. Where so indicated, alkyl groups can be optionally substituted. In substituent groups with multiple alkyl groups such as N(Ci_C6alky1)2, the alkyl groups may be the same or different.
As used herein, unless otherwise noted, "alkoxy" refers to groups of formula ¨
Oalkyl, wherein "alkyl" is as defined herein. Designated numbers of carbon atoms (e.g.
-0C1_C6) shall refer independently to the number of carbon atoms in the alkyl moiety of the alkoxy group, for example, but not limited to, from one to six carbon atoms or from one to three carbon atoms. Examples of alkoxy groups include, but are not limited to, methoxy, ethoxy, n-propoxy, iso-propoxy, n-butoxy, sec-butoxy, iso-butoxy, tert-butoxy, and the like. Where so indicated, alkoxy groups can be optionally substituted.
As used herein, unless otherwise noted, "aryl" whether used alone or part of another group refers to a carbocyclic fully unsaturated or partially unsaturated single or fused ring system. If the rings are fused, one of the rings must be fully unsaturated or partially unsaturated and the fused ring(s) may be fully saturated, partially unsaturated or fully unsaturated. The aryl group may be optionally substituted as defined herein. The term "aryl" embraces aromatic radicals such as phenyl, naphthyl, tetrahydronaphthyl, = PC72246 indanyl, biphenyl, benzo[b][1,4]oxazin-3(4H)-onyl, 2,3-dihydro-1H indenyl and 1,2,3,4-tetrahydronaphthalenyl.
As used herein, unless otherwise noted, "cycloalkyl" whether used alone or as part of another group, refers to a fully saturated hydrocarbon ring having from three to fourteen ring carbon atoms, for example, from four to seven; or from three to seven; or from three to six; or from three to five ring carbon atoms. Cycloalkyl groups can be monocyclic (e.g., cyclohexyl) or polycyclic (e.g., containing fused, bridged, and/or spiro ring systems), wherein the carbon atoms are located inside or outside of the ring system. Any suitable ring position of the cycloalkyl group can be covalently linked to the defined chemical structure. Where so indicated, cycloalkyl rings can be optionally substituted. Examples of cycloalkyl groups include, but are not limited to, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, cyclooctanyl, decalinyl.
The term "cycloalkyl" also includes carbocyclic rings which are bicyclic hydrocarbon rings, non-limiting examples of which include, bicyclo-[2.1.1jhexanyl, bicyclo[2.2.1]heptanyl, bicyclo[3. 1. 1Theptanyl, 1,3-d imethyl[2.2.1]heptan-2-yl, bicyclo[2.2.2]octanyl, and bicyclo[3.3.3]undecanyl.
As used herein, unless otherwise noted, the terms "haloalkyl" and "haloalkoxy"

are intended to include both branched and straight-chain saturated aliphatic "alkyl" or "alkoxy" groups respectively, wherein "alkyl" and "alkoxy" are as defined herein, having the specified number of carbon atoms and in which at least one hydrogen is replaced with a halogen atom. As used herein, the term "halogen atom" refers to F, Cl, Br and I.
Haloalkyl groups include perhaloalkyl groups, wherein all hydrogens of an alkyl group have been replaced with halogens (e.g., -CF3, -CF2CF3). In certain embodiments in which two or more hydrogen atoms are replaced by halogen atoms, the halogen atoms can be the same (e.g., CHF2, -CF3) or different (e.g., CF2CI). Where so indicated, haloalkyl or haloalkoxy groups can optionally be substituted with one or more substituents in addition to halogen. Examples of haloalkyl groups include, but are not limited to, fluoromethyl, dichloroethyl, trifluoromethyl, trichloromethyl, pentafluoroethyl, and pentachloroethyl groups.
As used herein, unless otherwise noted, the terms "heterocycly1" and "heterocycloalkyl" are used interchangeably and, whether used alone or as part of another group, are defined herein as referring to a group having one or more rings (e.g., 1, 2 or 3 rings) and having from 3 to 11 ring atoms (e.g. 3 to 6 ring atoms, 4 to 7 ring atoms, 4 to 5 ring atoms) wherein at least one ring atom, alternatively 1 to 5 ring atoms, alternatively 1 to 4 ring atoms, alternatively 1 to 3 ring atoms, alternatively one ring atom, alternatively two ring atoms, is a heteroatom, independently selected, unless indicated otherwise, from the group consisting of nitrogen (N), oxygen (0), and sulfur (S), and wherein the ring that includes the heteroatom is fully saturated.
Exemplary heterocyclyl groups have from 3 to 11 ring atoms, alternatively 4 to 7 ring atoms, alternatively 4 to 5 ring atoms, alternatively 3 to 6 ring atoms, of which, where chemically possible, from 1 to 5, alternatively 1 to 4, alternatively 1 to 3, alternatively 4, alternatively 3, alternatively 2, alternatively 1 ring atom, is a heteroatoms independently selected in each instance from, unless indicated otherwise, the group consisting of nitrogen (N), oxygen (0), or sulfur (S). In a group that has a heterocyclyl substituent, unless otherwise stated, the ring atom of the heterocyclyl substituent that is bound to the group may be one of the heteroatoms, or it may be a ring carbon atom, where the ring carbon atom may be in the same ring as the heteroatom(s), or the ring carbon may be in a different ring from the heteroatom(s). Where so indicated, the heterocyclyl substituent can be optionally further substituted with one or more group(s) or substituent(s), which group(s) or substituent(s) may be bound to the heteroatom(s) or may be bound to the ring carbon atom, where the ring carbon atom may be in the same ring as the at least one heteroatom or where the ring carbon atom may be in a different ring rom the heteroatom(s). Examples of monocyclic heterocyclyl groups include, but are not limited to, oxetanyl, diazirinyl, aziridinyl, urazolyl, azetidinyl, pyrazolidinyl, im id azolid i nyl, oxazolidinyl, isoxazolinyl, isoxazolyl, thiazolidinyl, isothiazolyl, isothiazolinyl oxathiazolidinonyl, oxazolidinonyl, hydantoinyl, tetrahydrofuranyl, pyrrolidinyl, morpholinyl, piperazinyl, piperidinyl, dihydropyranyl, tetrahydropyranyl, piperidin-2-onyl (valerolactam), 2,3,4,5-tetrahydro-1H-azepinyl, 2,3-dihydro-1H-indole, and 1,2,3,4-tetrahydro-quinoline.
As used herein, unless otherwise noted, the term "heteroaryl" whether used alone or as part of another group, is defined herein as a single or fused ring system having from five to eleven ring atoms (e.g. from five to ten ring atoms of from five to six ring atoms) wherein at least one ring atom, alternatively 2 ring atoms, alternatively 3 ring atoms, alternatively 4 ring atoms, in at least one ring is a heteroatom independently selected in each instance from, unless otherwise indicated, the group consisting of nitrogen (N), oxygen (0), and sulfur (S), and wherein further at least one of the rings comprising a heteroatom is fully unsaturated or partially unsaturated. In heteroaryl groups that include 2 or more fused rings, additional rings may bear one or more heteroatoms, may be a carbocycle (e.g., 6,7-Dihydro-5H-cyclopentapyrimidine) or may be aryl (e.g., benzofuranyl, benzo-thiophenyl, indolyl, indolinyl, tetrahydroquinolinyl, chromanyl, 1,4-dioxochromany1). In a group that has a heteroaryl substituent, unless otherwise indicated, the ring atom of the heteroaryl substituent that is bound to the group may be the at least one heteroatom, or it may be a ring carbon atom, where the ring carbon atom may be in the same ring as the at least one heteroatom or where the ring carbon may be in a different ring from the at least one heteroatom. Where so indicated, heteroaryl groups can be substituted.
If the heteroaryl substituent is substituted with a group or substituent, the group or substituent may be bound to the heteroatom, or it may be bound to a ring carbon atom, where the ring carbon atom may be in the same ring as the heteroatom(s), or where the ring carbon atom may be in a different ring from the heteroatom(s). Examples of monocyclic heteroaryl rings include, but are not limited to, 1,2,3,4-tetrazolyl, [1,2,3]triazolyl, [1,2,4]triazolyl, triazinyl, thiazol-2-yl, thiazol-4-yl, imidazol-1-yl, 1H-imidazol-2-yl, 1H-imidazol-4-yl, oxazolyl, isoxazolin-5-yl, furan-2-yl, furan-3-yl, thiophen-2-yl, thiophen-4-yl, pyrimidin-2-yl, pyrimidin-4-yl, pyrimidin-5-yl, pyridazinyl, pyrazinyl, pyridin-2-yl, pyridin-3-yl, and pyridin-4-y1 pyridinyl.
Examples of heteroaryl rings containing 2 or more fused rings include, but are not limited to, benzofuranyl, benzothiophenyl, benzoxazolyl, benzthiazolyl, benztriazolyl, cinnolinyl, naphthyridinyl, benzimidazolyl, aza-indolyl, aza-benzimidazolyl, phenanthridinyl, 7H-purinyl, 9H-purinyl, 5H-pyrrolo[3,2-d]pyrimidinyl, 7H-pyrrolo[2,3-d]pyrimidinyl, pyrido[2,3-d]pyrimidinyl, 2-phenylbenzo[d]thiazolyl, 1H-indolyl, 4,5,6,7-tetrahydro-1-H-indolyl, quinoxalinyl, 5-methylquinoxalinyl, quinazolinyl, quinolinyl, and isoquinolinyl. The term "heteroaryl" also includes pyridyl N-oxides and groups containing a pyridine N-oxide ring.
As used herein, unless otherwise stated, the term "amino" refers to ¨N H2.
As used herein, unless otherwise stated, the term "alkylamino" refers to ¨
N(H)alkyl, the term "alkyl" having already been defined herein. Examples of alkylamino substituents include, but are not limited to, methylamino, ethylamino, and propylamino.

= = P072246 As used herein, unless otherwise stated, the term "dialkylamino" refers to ¨
N(alkyl)2 where the two alkyls may be the same or different and where the term "alkyl"
has already been defined herein. Examples of dialkylamino substituents include, but are not limited to, dimethylamino, diethylamino, ethylmethylamino, and dipropylamino.
5 As used herein, unless otherwise stated, the term "amido" refers to ¨C(=0)NH2.
As used herein, unless otherwise stated, the term "halogen" or "halogen atom"
refers to the group consisting of fluorine (which may be depicted as ¨F), chlorine (which may be depicted as ¨Cl), bromine (which may be depicted as ¨Br), or iodine (which may be depicted as ¨I).

As used herein, unless otherwise stated, the terms "hydroxy" and "hydroxyl"
are used interchangeably and as used herein mean an -OH group. As used herein, unless otherwise noted, the terms "hydroxyalkyl" and "hydroxyalkoxy" are intended to include both branched and straight-chain saturated aliphatic "alkyl" or "alkoxy"
groups respectively, wherein "alkyl" and "alkoxy" are as defined herein, having the specified number of carbon atoms and in which at least one hydrogen is replaced with a ¨OH
group. Where so indicated, hydroxyalkyl and hydroxyalkoxy groups can optionally be substituted with one or more substituents in addition to -OH. Examples of hydroxyalkyl groups include, but are not limited to, CH2OH, CH2CH2OH, CH2(OH)CH2OH.
As used herein, unless otherwise stated, the term "oxo" =0.
20 As used herein, unless otherwise stated, the term "carbonyl" refers to 0=0.
As used herein, unless otherwise stated, the term "carboxy" refers to ¨CO2H.
As used herein, unless otherwise stated, the term sulfonyl refers to -SO2-.
As used herein, the term "substituted" is used throughout the specification.
The term "substituted" is defined herein as a moiety, whether acyclic or cyclic, which has one or more (e.g. 1-10) hydrogen atoms replaced by a substituent as defined herein below. Substituents include those that are capable of replacing one or two hydrogen atoms of a single moiety at a time, and also those that can replace two hydrogen atoms on two adjacent carbons to form said substituent. For example, substituents that replace single hydrogen atoms include, but are not limited to, halogen, hydroxy, and the like. A two hydrogen atom replacement includes, but is not limited to, carbonyl, oximino, and the like. Substituents that replace two hydrogen atoms from adjacent ' = PC72246 carbon atoms include, but are not limited to, epoxy, and the like. When a moiety is described as "substituted" any number of its hydrogen atoms can be replaced, as described above. For example, difluoromethyl is a substituted C1 alkyl;
trifluoromethyl is a substituted C1 alkyl; 4-hydroxyphenyl is a substituted aryl ring; (N,N-dimethy1-5-amino)octanyl is a substituted C8 alkyl; 3-guanidinopropyl is a substituted C3 alkyl; and 2-carboxy-3-fluoropyridinyl is a substituted heteroaryl.
A multi-moiety substituent is bound through the atom indicated by "-". To illustrate this the term "-0C1-C3hydroxyalkyl" is an OC1-C3alkyl group substituted by a hydroxy group. Further, any carbon number pre-fix attached to a multi-moiety substituent only applies to the moiety it immediately precedes. To illustrate, the term "cycloalkyl(Ci-C4)alkyl" contains two moieties: alkyl and cycloalkyl. The (C1-C4) pre-fix on the cycloalkyl(C1-C4)alkyl means that the alkyl moiety of the alkylcycloalkyl contains from 1 to 4 carbon atoms, the (C1-C4) pre-fix does not describe the cycloalkyl moiety.
If a group of substituents are collectively described as being optionally substituted by one or more of a list of substituents, the group may include (1) unsubstitutable substituents, (2) substitutable substituents that are not substituted by the optional substituents, and / or (3) substitutable substituents that are substituted by one or more of the optional substituents.
If a substituent is described such that it "may be substituted" or as being "optionally substituted" with up to a particular number of non-hydrogen substituents, that substituent may be either (1) not substituted; or (2) substituted by up to that particular number of non-hydrogen substituents or by up to the maximum number of substitutable positions on the substituents, whichever is less. Thus, for example, if a substituent is described as a heteroaryl optionally substituted with one, two or three substituents, then any heteroaryl with less than three substitutable positions would be optionally substituted by up to only as many non-hydrogen substituents as the heteroaryl has substitutable positions. To illustrate, tetrazolyl (which has only one substitutable position) would be optionally substituted with up to one non-hydrogen substituent.
At various places in the present specification, substituents of compounds are disclosed in groups or in ranges. It is specifically intended that the description include each and every individual sub-combination of the members of such groups and ranges.
For example, the term "C1_6 alkyl" is specifically intended to individually disclose C1, C2, = ' ' PC72246 C3, C4, 05, 06, 01-06, 01-05, Ci-04, Ci-C3, C1-C2, C2-C6, 02-05, 02-04, C2-C3, 03-06, 03-06, 03-04, 04-06, C4-05, and 05-06 alkyl. For example, the term "Ci_3 alkyl"
is specifically intended to individually disclose Cl, 02, 03, C1-C3, C1-C2, and 02-C3 alkyl.
Still further examples of BET-family bromodomain inhibitors that may be used in the present invention include those compounds generically or specifically disclosed in the following patent application publications: W02015/004075; W02015/004534;
W02015/004533; W02015/002754; US2015011540; W02014/206150; W014206345;
W02014/202578; W02014/191894; W02014/191906;
W02014/191911;
US2014349990; US2014336190; W02014/182929; W02014/173241; EP2792355;
W02014/170350; US2014296246; W02014/160873;
US2014296229;
W02014/154760; US2014296230; W02014/154762;
W02014/152029;
US2014275030; W02014/143768; W02014/145051; US2014275026; US2014275079;
W02014/139324; W02014/134232; W02014/134267;
W02014/128070;
W02014/128067; W02014/128111; W02014/128655; W02014/095774;
W02014/095775; US2014162971; 0N103833671; US2014140956; US2014142102;
US2014142098; W02014/076237; US2014135336;
W02014/048945;
W02014/026997; US2013331382; W02013/184878;
W02013/184876;
W02013/155695; US2013281450; W02013/158952;
W02013/097052;
W02013/097601; US2013079335; W02013/033268; W02013/033269;
W02013/030150; W02013/027168; GB201114103;
W02013/024104;
W02012/151512; W02012/150234; W02012/143413;
W02012/143416;
W02012/143415; W02012/116170; W02012/075383;
W02012/075456;
W02011/161031; GB200919426; W02011/054844;
W02011/054845;
W02011/054553; W02011/054843; W02011/054848; W02011/054846;
W02009/084693; EP2239264; US2010286127 or pharmaceutically acceptable salts thereof, all of which are incorporated by reference herein in their entirety for all purposes.
In one embodiment of the present invention the BET-family bromodomain inhibitor is I-BET-762, or a pharmaceutically acceptable salt thereof.
In one embodiment of the present invention the BET-family bromodomain inhibitor is JO-I, or a pharmaceutically acceptable salt thereof.

' PC72246 In one embodiment of the present invention the BET-family bromodomain inhibitor is JQ-1(+), or a pharmaceutically acceptable salt thereof.
In one embodiment of the present invention BET-family bromodomain inhibitor is CPI-203, or a pharmaceutically acceptable salt thereof.
In one embodiment of the present invention the BET-family bromodomain inhibitor is OTX-015, or a pharmaceutically acceptable salt thereof.
In one embodiment of the present invention the BET-family bromodomain inhibitor is GW-841819X, or a pharmaceutically acceptable salt thereof.
In one embodiment of the present invention the BET-family bromodomain inhibitor is CP-0610, or a pharmaceutically acceptable salt thereof.
In one embodiment of the present invention the BET-family bromodomain inhibitor is CPI-232, or a pharmaceutically acceptable salt thereof.
In one embodiment of the present invention the BET-family bromodomain inhibitor is BET-BAY-002, or a pharmaceutically acceptable salt thereof.
In one embodiment of the present invention the BET-family bromodomain inhibitor is RVX-208, or a pharmaceutically acceptable salt thereof.
In one embodiment of the present invention the BET-family bromodomain inhibitor is I-BET-726, or a pharmaceutically acceptable salt thereof.
In one embodiment of the present invention the BET-family bromodomain inhibitor is SRX-2523, or a pharmaceutically acceptable salt thereof.
In one embodiment of the present invention the BET-family bromodomain inhibitor is BMS-986158, or a pharmaceutically acceptable salt thereof.
In In one embodiment of the present invention the BET-family bromodomain inhibitor is RVX-297, or a pharmaceutically acceptable salt thereof.
In one embodiment of the present invention the BET-family bromodomain inhibitor is RVS-2135, or a pharmaceutically acceptable salt thereof.
In one embodiment of the present invention the BET-family bromodomain inhibitor is ZEN-3365, or a pharmaceutically acceptable salt thereof.

, PC72246 In one embodiment of the present invention the BET-family bromodomain inhibitor is ZEN-3118, or a pharmaceutically acceptable salt thereof.
In one embodiment of the present invention the BET-family bromodomain inhibitor is KM-601, or a pharmaceutically acceptable salt thereof.
In one embodiment of the present invention the BET-family bromodomain inhibitor is SF-2535, or a pharmaceutically acceptable salt thereof.
In one embodiment of the present invention the BET-family bromodomain inhibitor is AU-004, or a pharmaceutically acceptable salt thereof.
In one embodiment of the present invention the BET-family bromodomain inhibitor is ABBV-075, or a pharmaceutically acceptable salt thereof.
In one embodiment of the present invention the BET-family bromodomain inhibitor is TEN-010, or a pharmaceutically acceptable salt thereof.
In one embodiment of the present invention the BET-family bromodomain inhibitor is BAY1238097, or a pharmaceutically acceptable salt thereof.
In one embodiment of the present invention the BET-family bromodomain inhibitor is N-(2-hydroxy-3-methylquinolin-6-yl)piperidine-1-sulfonamide, or a pharmaceutically acceptable salt thereof.
In one embodiment of the present invention the BET-family bromodomain inhibitor is N44-(2,4-difluorophenoxy)-3-(6-methyl-7-oxo-6,7-dihydro-1H-pyrrolo[2,3-c]pyridin- 4-yl)phenyl]ethanesulfonamide, or a pharmaceutically acceptable salt thereof.
In one embodiment of the present invention the BET-family bromodomain inhibitor is N-{64methyl(propanoyl)aminol-3-oxo-4-[(1S)-1-phenylpropyl]-3,4-dihydropyrido[2,3 b]pyrazin-2-yll-beta-alanine, or a pharmaceutically acceptable salt thereof.
In some embodiments of the present invention the BET-family bromodomain inhibitor is selected from the group consisting of:
I-BET762;
JQ-1;
JQ-1(+);

. PC72246 CPI-267203;
OTX-015;
GW-841819X;
CP-0610;
5 CPI-232;
BET-BAY-002;
1-BET-151;
RVX-208;
I-BET-726;
10 SRX-2523;
N-(2-hydroxy-3-methylquinolin-6-yl)piperidine-1-sulfonamide;
N-[4-(2,4-difluorophenoxy)-3-(6-methy1-7-oxo-6,7-dihydro-1H-pyrrolo[2,3-c]pyridin- 4-yl)phenyliethanesulfonamide; and N-16-[methyl(propanoyDamino]-3-oxo-4-[(1S)-1-phenylpropyl]-3,4-15 dihydropyrido[2,3 b]pyrazin-2-yll-beta-alanine or a pharmaceutically acceptable salt thereof.
In some embodiments of the present invention the BET-family bromodomain inhibitor is selected from the group consisting of:
I-BET762;
20 JQ-1;
JQ-1(+);
RVX-208;
1-BET-151;
N-(2-hydroxy-3-methylquinolin-6-yl)piperidine-1-sulfonamide;
25 N-[4-(2,4-difluorophenoxy)-3-(6-methy1-7-oxo-6,7-dihydro-1H-pyrrolo[2,3-c]pyridin- 4-yl)phenyllethanesulfonamide; and = a' PC72246 N-{6-[methyl(propanoyl)amino]-3-oxo-4-[(1S)-1-phenylpropyl]-3,4-dihydropyrido[2,3 b]pyrazin-2-yI}-beta-alanine or a pharmaceutically acceptable salt thereof.
Compounds useful for the present invention may be in an unsolvated or solvated forms. The term "solvate" as used herein means a physical association of a compound with one or more solvent molecules, whether organic or inorganic, including water ('hydrate'). When the solvent or water is tightly bound, the complex will have a well-defined stoichiometry independent of humidity. When, however, the solvent or water is weakly bound, as in channel solvates and hygroscopic compounds, the water/solvent content will be dependent on humidity and drying conditions. In such cases, non-stoichiometry will be the norm.
Compounds useful for the present invention may be used in the form of salts derived from inorganic or organic acids. Depending on the particular compound, a salt of the compound may be advantageous due to one or more of the salt's physical properties, which may include enhanced pharmaceutical stability in differing temperatures and humidities, or a desirable solubility in water or oil. In some instances, a salt of a compound also may be used as an aid in the isolation, purification, and/or resolution of the compound.
Where a salt is intended to be administered to a patient (as opposed to, for example, being used in an in vitro context), the salt preferably is pharmaceutically acceptable. The term "pharmaceutically acceptable salt" refers to a salt prepared by combining a compound for use in the invention with an acid whose anion, or a base whose cation, is generally considered suitable for human consumption.
Pharmaceutically acceptable salts may be particularly useful in the present invention because of their potentially greater aqueous solubility relative to the parent compound.
Salts encompassed within the term "pharmaceutically acceptable salts" refer to non-toxic salts of the compounds of this invention which may be prepared by reacting the free base with a suitable organic or inorganic acid.
When possible, suitable pharmaceutically acceptable acid addition salts of the compounds for use in the present invention may include those derived from inorganic acids, such as hydrochloric, hydrobromic, hydrofluoric, boric, fluoroboric, phosphoric, metaphosphoric, nitric, carbonic, sulfonic, and sulfuric acids, and organic acids such as = - PC72246 acetic, benzenesulfonic, benzoic, citric, ethanesulfonic, funnaric, gluconic, glycolic, isothionic, lactic, lactobionic, maleic, malic, methanesulfonic, trifluoromethanesulfonic, succinic, toluenesulfonic, tartaric, and trifluoroacetic acids. Suitable organic acids may include but are not limited to aliphatic, cycloaliphatic, aromatic, araliphatic, heterocyclic, carboxylic, and sulfonic classes of organic acids.
Specific examples of suitable organic acids may include but are not limited to acetate, trifluoroacetate, formate, propionate, succinate, glycolate, gluconate, digluconate, lactate, malate, tartaric acid, citrate, ascorbate, glucuronate, maleate, fumarate, pyruvate, aspartate, glutamate, benzoate, anthranilic acid, stearate, salicylate, p-hydroxybenzoate, phenylacetate, mandelate, embonate (pamoate), methanesulfonate, ethanesulfonate, benzenesulfonate, pantothenate, toluenesulfonate, 2-hydroxyethanesulfonate, sufanilate, cyclohexylaminosulfonate, algenic acid, .beta.-hydroxybutyric acid, galactarate, galacturonate, adipate, alginate, butyrate, camphorate, camphorsulfonate, cyclopentanepropionate, dodecylsulfate, glycoheptanoate, glycerophosphate, heptanoate, hexanoate, nicotinate, 2-naphthalesulfonate, oxalate, palmoate, pectinate, 3-phenylpropionate, picrate, pivalate, thiocyanate, and undecanoate.
Furthermore, where the compounds for use in the present invention carry an acidic moiety, suitable pharmaceutically acceptable salts thereof may include alkali metal salts, i.e., sodium or potassium salts; alkaline earth metal salts, e.g., calcium or magnesium salts; and salts formed with suitable organic ligands, e.g., quaternary ammonium salts. In another embodiment, base salts are formed from bases which form non-toxic salts, which may include aluminum, arginine, benzathine, choline, diethylamine, diolamine, glycine, lysine, meglumine, olamine, tromethamine and zinc salts.
Organic salts may be made from secondary, tertiary or quaternary amine salts, such as tromethamine, diethylamine, N,N'-benzylethylenediamine, chloroprocaine, choline, diethanolamine, ethylenediamine, meglumine (N-methylglucamine), and procaine. Basic nitrogen-containing groups may be quaternized with agents such as lower alkyl (C1-C 6) halides (e.g., methyl, ethyl, propyl, and butyl chlorides, bromides, and iodides), dialkyl sulfates (i.e., dinnethyl, diethyl, dibutyl, and diamyl sulfates), long chain halides (i.e., decyl, lauryl, myristyl, and stearyl chlorides, bromides, ' PC72246 and iodides), arylalkyl halides (i.e., benzyl and phenethyl bromides), and others.
Compounds for use in the present invention may also be in the form of hemisalts of acids and bases, for example, hemisulphate and hemicalcium salts.
Compounds for use in the present invention may also be in the form of complexes. Complexes, such as clathrates, drug-host inclusion complexes wherein, in contrast to the aforementioned solvates, the drug and host are present in stoichiometric or non-stoichiometric amounts may be used. Also, complexes of the drug containing two or more organic and/or inorganic components which may be in stoichiometric or non-stoichiometric amounts may be used. The resulting complexes may be ionised, partially ionised, or non-ionised. For a review of such complexes, see J Pharm Sci, 64 (8), 1269-1288 by Haleblian (August 1975).
Compounds for use in the present invention may also be in the form of a prodrug.
The term "prodrug" as used herein, refers to a substantially pharmacologically inactive derivative of a parent "drug" molecule that requires biotransformation (e.g., either spontaneous or enzymatic) within the target physiological system to release or convert the prodrug into the active drug. Prodrugs are designed to overcome problems which may include problems associated with stability, toxicity, lack of specificity, or limited bioavailability. Exemplary prodrugs comprise an active drug molecule itself and a chemical masking group (e.g., a group that reversibly suppresses the activity of the drug). Some preferred prodrugs may be variations or derivatives of compounds that have groups cleavable under metabolic conditions. Exemplary prodrugs become pharmacologically active in vivo or in vitro when they undergo solvolysis under physiological conditions or undergo enzymatic degradation or other biochemical transformation (e.g., phosphorylation, hydrogenation, dehydrogenation, glycosylation).
Prodrugs may offer advantages of solubility, tissue compatibility, or delayed release in the mammalian organism. (See e.g., Bundgard, Design of Prodrugs, pp. 7-9, 21-24, Elsevier, Amsterdam (1985); and Silverman, The Organic Chemistry of Drug Design and Drug Action, pp. 352-401, Academic Press, San Diego, CA (1992)). Common prodrugs include acid derivatives such as esters prepared by reaction of parent acids with a suitable alcohol (e.g., a lower alkanol), amides prepared by reaction of the parent acid compound with an amine, or basic groups reacted to form an acylated base derivative (e.g., a lower alkylamide).

Compounds for use in the present invention may also be in the form of isotopically labelled derivatives of the BET-family bromodomain inhibitor wherein one or more atoms are replaced by atoms having the same atomic number, but an atomic mass or mass number different from the atomic mass or mass number usually found in nature. Examples of isotopes that may be suitable for inclusion in the compounds for use in the present invention include isotopes of hydrogen, such as 2H and 3H, carbon, such as 11C, 13C and 14C, chlorine, such as 36CI, fluorine, such as 18F, iodine, such as 1231 and 1251, nitrogen, such as 13N and 15N, oxygen, such as 150, 170 and 180, phosphorus, such as 32P, and sulphur, such as 36S. Substitution with heavier isotopes such as deuterium, i.e. 2H, may afford certain advantages resulting from potentially greater metabolic stability, for example, potentially increased in vivo half-life or potentially reduced dosage requirements, and hence may be preferred in some circumstances. Substitution with positron emitting isotopes, such as 11C, I" 150 and 13N, may be useful in Positron Emission Topography (PET) studies for examining substrate receptor occupancy. Isotopically-labeled compounds can generally be prepared by conventional techniques known to those skilled in the art or by processes analogous to those described.
Pharmaceutical Dosage Forms For the pharmaceutical uses of the present invention, BET-family bromodomain inhibitors, and pharmaceutically acceptable salts thereof, will usually be formulated in a pharmaceutically acceptable composition.
Methods of formulation of such pharmaceutically acceptable compositions are well known in the art and are disclosed, for example, in Remington: The Science and Practice of Pharmacy, Mack Publishing Company, Easton, Pa., 21st Edition (2005), incorporated herein by reference in its entirety for all purposes.
Pharmaceutical compositions suitable for use in the present invention may be in the form of sterile, non-pyrogenic liquid solutions or suspensions, coated capsules, suppositories, lyophilized powders, transdermal patches or other forms known in the art.
Injectable preparations, for example, sterile injectable aqueous or oleaginous suspensions may be formulated according to the known art using suitable dispersing or wetting agents and suspending agents. The sterile injectable preparation may also be a sterile injectable solution, suspension or emulsion in a nontoxic parenterally acceptable diluent or solvent.
In addition, sterile, fixed oils may be conventionally employed as a solvent or suspending medium. For this purpose any bland fixed oil may be employed including 5 synthetic mono- or di-glycerides. In addition, fatty acids such as oleic acid may find use in the preparation of injectables. The injectable formulations may be sterilized, for example, by filtration through a bacterial-retaining filter, or by incorporating sterilizing agents in the form of sterile solid compositions which may be dissolved or dispersed in sterile water or other sterile injectable medium prior to use.
10 Formulations may be provided comprising crystalline forms of the pharmaceutically active agent for slow absorption from subcutaneous or intramuscular injection. Additionally, delayed absorption of a parenterally administered drug form may be accomplished by dissolving or suspending the compounds in an oil vehicle.
Injectable depot forms may be made by forming microencapsule matrices of the drug in 15 biodegradable polymers such as polylactide-polyglycolide. Depending upon the ratio of drug to polymer and the nature of the particular polymer employed, the rate of drug release may be controlled. Examples of other biodegradable polymers include poly(orthoesters) and poly(anhydrides). Depot injectable formulations may also be prepared by entrapping the drug in liposomes or microemulsions, which are compatible 20 with body tissues.
Solid dosage forms intended for oral administration may include capsules, tablets, pills, powders, and granules. In such solid dosage forms, the active compound may be mixed with at least one inert, pharmaceutically acceptable excipient or carrier such as sodium citrate or dicalcium phosphate and/or a) fillers or extenders such as 25 starches, lactose, sucrose, glucose, mannitol, and silicic acid, b) binders such as, for example, carboxymethylcellulose, alginates, gelatin, polyvinylpyrrolidinone, sucrose, and acacia, c) humectants such as glycerol, d) disintegrating agents such as agar-agar, calcium carbonate, potato or tapioca starch, alginic acid, certain silicates, and sodium carbonate, e) solution retarding agents such as paraffin, f) absorption accelerators such 30 as quaternary ammonium compounds, g) wetting agents such as, for example, acetyl alcohol and glycerol monostearate, h) absorbents such as kaolin and bentonite clay, and i) lubricants such as talc, calcium stearate, magnesium stearate, solid polyethylene ' PC72246 glycols, sodium lauryl sulfate, and mixtures thereof. In the case of capsules, tablets and pills, the dosage form may also comprise buffering agents.
Solid compositions of a similar type may also be employed as fillers in soft and hard-filled gelatin capsules using such excipients as lactose or milk sugar as well as high molecular weight polyethylene glycols and the like.
The solid dosage forms of tablets, capsules, pills, and granules may be prepared with coatings and shells such as enteric coatings and other coatings well known in the pharmaceutical formulating art. They may optionally contain opacifying agents and may also be of a composition that they release the active ingredient(s) only, or preferentially, in a certain part of the intestinal tract, optionally, in a delayed manner.
Examples of embedding compositions that may be used include polymeric substances and waxes.
Alternative formulations may include those where the pharmaceutically active agent is in micro-encapsulated form with one or more excipients as noted above. The solid dosage forms of tablets, capsules, pills, and granules may be prepared with coatings and shells such as enteric coatings, release controlling coatings and other coatings well known in the pharmaceutical formulating art. In such solid dosage forms the active compound may be admixed with at least one inert diluent such as sucrose, lactose or starch. Such dosage forms may also comprise, as is normal practice, additional substances other than inert diluents, e.g., tableting lubricants and other tableting aids such a magnesium stearate and microcrystalline cellulose. In the case of capsules, tablets and pills, the dosage forms may also comprise buffering agents. They may optionally contain opacifying agents and may also be of a composition that they release the active ingredient(s) only, or preferentially, in a certain part of the intestinal tract, optionally, in a delayed manner. Examples of embedding compositions that may be used include polymeric substances and waxes.
Liquid dosage forms intended for oral administration may include pharmaceutically acceptable emulsions, microemulsions, solutions, suspensions, syrups and elixirs. In addition to the active compounds, the liquid dosage forms may contain inert diluents commonly used in the art such as, for example, water or other solvents, solubilizing agents and emulsifiers such as ethyl alcohol, isopropyl alcohol, ethyl carbonate, Et0Ac, benzyl alcohol, benzyl benzoate, propylene glycol, 1,3-butylene glycol, dimethylformamide, oils (in particular, cottonseed, groundnut, corn, germ, olive, ' PC72246 castor, and sesame oils), glycerol, tetrahydrofurfuryl alcohol, polyethylene glycols and fatty acid esters of sorbitan, and mixtures thereof. Besides inert diluents, the oral compositions may also include adjuvants such as wetting agents, emulsifying and suspending agents, sweetening, flavoring, and perfuming agents.
Dosage forms intended for topical or transdermal administration of a compound of this invention may include ointments, pastes, creams, lotions, gels, powders, solutions, sprays, inhalants or patches. The pharmaceutically active agent may be admixed under sterile conditions with a pharmaceutically acceptable carrier and any needed preservatives or buffers as may be required. Ophthalmic formulations, ear drops, and the like are also contemplated.
Compositions may also be formulated for delivery as a liquid aerosol or inhalable dry powder. Liquid aerosol formulations may be nebulized predominantly into particle sizes that can be delivered to the terminal and respiratory bronchioles.
The phrase "pharmaceutically acceptable carrier" as used herein means a pharmaceutically-acceptable material, composition or vehicle, such as a liquid or solid filler, diluent, excipient, solvent or encapsulating material, involved in carrying or transporting the pharmacologically active agent from one organ, or portion of the body, to another organ, or portion of the body. Each carrier must be "acceptable" in the sense of being compatible with the other ingredients of the formulation and not injurious to a patient. Some examples of materials which may serve as pharmaceutically-acceptable carriers include: (1) sugars, such as lactose, glucose and sucrose; (2) starches, such as corn starch and potato starch; (3) cellulose, and its derivatives, such as sodium carboxymethyl cellulose, ethyl cellulose and cellulose acetate; (4) powdered tragacanth;
(5) malt; (6) gelatin; (7) talc; (8) excipients, such as cocoa butter and suppository waxes; (9) oils, such as peanut oil, cottonseed oil, safflower oil, sesame oil, olive oil, corn oil and soybean oil; (10) glycols, such as propylene glycol; (11) polyols, such as glycerin, sorbitol, mannitol and polyethylene glycol; (12) esters, such as ethyl oleate and ethyl laurate; (13) agar; (14) buffering agents, such as magnesium hydroxide and aluminum hydroxide; (15) alginic acid; (16) pyrogen-free water; (17) isotonic saline; (18) Ringer's solution; (19) ethyl alcohol; (20) phosphate buffer solutions; and (21) other non-toxic compatible substances employed in pharmaceutical formulations. A
pharmaceutically acceptable carrier should not cause significant irritation to an ' PC72246 organism and does not abrogate the biological activity and properties of the pharmacologically active agent.
An "excipient" refers to an inert substance added to a pharmacological composition to further facilitate administration of a pharmacologically active agent.
Examples of excipients may include but are not limited to calcium carbonate, calcium phosphate, various sugars and types of starch, cellulose derivatives, gelatin, vegetable oils and polyethylene glycols.
Dosing and Administration The compounds for use in the invention described herein may be administered orally, parenterally, sublingually, by aerosolization or inhalation spray, rectally, intracisternally, intravaginally, intraperitoneally, bucally, intrathecally or topically in dosage unit formulations containing conventional nontoxic pharmaceutically acceptable carriers, adjuvants, and vehicles as desired. The term parenteral as used herein includes subcutaneous injection, intravenous injection, intramuscular injection, intrasternal injection, or infusion techniques. Topical administration may also involve the use of transdermal administration such as transdermal patches or ionophoresis devices.
Effective amounts of the compounds for use in the invention generally include any amount sufficient to detectably modulate BET-family bromodomain activity, or, in some embodiments, to alleviate symptoms associated with diseases associated with BET-family bromodomain activity or susceptible to BET-family bromodomain activity modulation.
The specific dose of compound used according to the aspects of the invention to obtain therapeutic and/or prophylactic effect will vary depending upon the host treated and the particular mode of administration. It will be understood, however, that the specific dose level for any particular subject will depend upon a variety of factors including, for example, the specific compound administered, the route of administration, the condition being treated, and the individual being treated, including the age, body weight, general health, sex, diet, of the individual, the time of administration, route of administration, rate of excretion, drug combination, and the severity of the particular disease undergoing therapy. The therapeutically effective amount for a given situation can be readily determined by routine experimentation and is within the skill of one in the ' PC72246 art. A typical daily dose (which may be administered in single or divided doses) may contain a dosage level of from about 0.01 mg/kg to about 50-100 mg/kg of body weight of an active compound for use in the invention. Preferred daily doses may generally be from about 0.05 mg/kg to about 20 mg/kg and may ideally be from about 0.1 mg/kg to about 10 mg/kg. Factors such as clearance rate, half-life and maximum tolerated dose (MTD) can determine these using standard procedures.
Furthermore, it will be understood by those skilled in the art that the dosing may be adjusted to balance treatment efficacy whilst minimizing patient adverse events and thereby improving the safety profile. Such dosing adjustments would be made by taking into account routine parameters such as the half life of frataxin and drug exposure.
In certain pharmaceutical use embodiments, the BET-family bromodomain inhibitor, or pharmaceutically acceptable salt thereof, may be used in combination with a second pharmaceutically active ingredient, or pharmaceutically acceptable salt thereof.
As used herein, the terms "co-administration", "co-administered", "a combination of" or "in combination with", refers to a combination of a compound for use in the invention (e.g., a compound of Formula (I)) and one or more other pharmaceutically active ingredient, or a pharmaceutically acceptable salt thereof, and includes the following:
a. simultaneous use of such a combination of a BET-family bromodomain inhibitor and a further pharmaceutically active agent in a patient in need of treatment, when such components are formulated together into a single dosage form which releases said components at substantially the same time to said patient, b. substantially simultaneous use of such a combination of a BET-family bromodomaininhibitor and a further pharmaceutically active agent in a patient in need of treatment, when such components are formulated apart from each other into separate dosage forms which are used at substantially the same time by said patient, whereupon said components are released at substantially the same time to said patient, c. sequential use of such a combination of a BET-family bromodomain inhibitor and a further pharmaceutically active agent in a patient in need of treatment, when such components are formulated apart from each other into separate dosage forms which are used at consecutive times by said patient with a significant time = , PC72246 interval between each administration, whereupon said components are released at substantially different times to said patient; and, d. sequential use of such a combination of a BET-family bromodomain inhibitor and a further pharmaceutically active agent in a patient in need of treatment, when 5 such components are formulated together into a single dosage form which releases said components in a controlled manner.
The compounds for use in the present invention may be used with one or more therapeutic agents selected from the group consisting of:
(I) an antihypertensive agent. Examples of suitable anti-hypertensive agents may 10 include: alpha adrenergic blockers; beta adrenergic blockers; calcium channel blockers (e.g., diltiazem, verapamil, nifedipine and amlodipine); vasodilators (e.g., hydralazine), diruetics (e.g., chlorothiazide, hydrochlorothiazide, flumethiazide, hydroflumethiazide, bendroflumethiazide, methylchlorothiazide, trichloromethiazide, polythiazide, benzthiazide, ethacrynic acid tricrynafen, 15 chlorthalidone, torsemide, furosemide, musolimine, bumetanide, triamtrenene, amiloride, spironolactone); renin inhibitors; ACE inhibitors (e.g., captopril, zofenopril, fosinopril, enalapril, ceranopril, cilazopril, delapril, pentopril, quinapril, ramipril, lisinopril); AT-1 receptor antagonists (e.g., losartan, irbesartan, valsartan); ET receptor antagonists (e.g., sitaxsentan, atrsentan and compounds 20 disclosed in U.S. Patent Nos. 5,612,359 and 6,043,265); Dual ET/All antagonist (e.g., compounds disclosed in WO 00/01389); neutral endopeptidase (NEP) inhibitors; vasopepsidase inhibitors (dual NEP-ACE inhibitors) (e.g., gemopatrilat and nitrates). An exemplary antianginal agent that may be used is ivabradine;
(ii) a calcium channel blocker (L-type or T-type) or a cardiac glycoside.
Examples of 25 suitable calcium channel blockers may include diltiazem, verapamil, nifedipine and amlodipine and mybefradil. Examples of suitable cardiac glycosides may include digitalis and ouabain;
(iii) a diuretic. Examples of suitable diuretics may include (a) loop diuretics such as furosemide (such as LASIXTm), torsemide (such as DEMADEXTm), bemetanide 30 (such as BUMEXTm), and ethacrynic acid (such as EDECRINTm); (b) thiazide-type diuretics such as chlorothiazide (such as DIURILTM, ESIDRIXTM or HYDRODIURILTm), hydrochlorothiazide (such as MICROZIDETM or ORETICTm), benzthiazide, hydroflumethiazide (such as SALURONTm), bendroflumethiazide, methychlorthiazide, polythiazide, trichlormethiazide, and indapamide (such as LOZOLTm); (c) phthalimidine-type diuretics such as chlorthalidone (such as HYGROTONTm), and metolazone (such as ZAROXOLYNTm); (d) quinazoline-type diuretics such as quinethazone; and (e) potassium-sparing diuretics such as triamterene (such as DYRENIUMTm), and amiloride (such as MIDAMORTm or MODURETICTm). In another embodiment, a compound for use in the present invention may be co-administered with a loop diuretic.
In still another embodiment, the loop diuretic may be selected from furosemide and torsemide.
In still another embodiment, one or more compounds for use in the present invention may be co-administered with furosemide. In still another embodiment, one or more compounds for use in the present invention may be co-administered with torsemide which may optionally be a controlled or modified release form of torsemide. The diuretic may be a thiazide-type diuretic selected from the group consisting of chlorothiazide and hydrochlorothiazide. In still another embodiment, one or more compounds for use in the invention may be co-administered with chlorothiazide. In still another embodiment, one or more compounds for use in the present invention may be co-administered with hydrochlorothiazide. In another embodiment, one or more compounds for use in the invention may be co-administered with a phthalimidine-type diuretic. In still another embodiment, the phthalimidine-type diuretic may be chlorthalidone;
(iv) an anti-diabetic agent, particularly type 2 anti-diabetic agent.
Examples of suitable anti-diabetic agents may include (e.g. insulins, metfomin, DPPIV
inhibitors, GLP-1 agonists, analogues and mimetics, SGLT1 and SGLT2 inhibitors) Suitable anti-diabetic agents may include an acetyl-CoA
carboxylase-(ACC) inhibitor such as those described in W02009144554, W02003072197, W02009144555 and W02008065508, a diacylglycerol 0-acyltransferase 1 (DGAT-1) inhibitor, such as those described in W009016462 or W02010086820, AZD7687 or LCQ908, diacylglycerol 0-acyltransferase 2 (DGAT-2) inhibitor, monoacylglycerol 0-acyltransferase inhibitors, a PDE10 inhibitor, an AMPK
activator, a sulfonylurea (e.g., acetohexamide, chlorpropamide, diabinese, glibenclamide, glipizide, glyburide, glimepiride, gliclazide, glipentide, gliquidone, glisolamide, tolazamide, and tolbutamide), a meglitinide, an a-amylase inhibitor (e.g., tendamistat, trestatin and AL-3688), an a-glucoside hydrolase inhibitor " PC72246 I =

(e.g., acarbose), an a-glucosidase inhibitor (e.g., adiposine, camiglibose, emiglitate, miglitol, voglibose, pradimicin-Q, and salbostatin), a PPARy agonist (e.g., balaglitazone, ciglitazone, darglitazone, englitazone, isaglitazone, pioglitazone and rosiglitazone), a PPAR a/y agonist (e.g., CLX-0940, GW-1536, GW-1929, GW-2433, KRP-297, L-796449, LR-90, MK-0767 and SB-219994), a biguanide (e.g., metformin), a glucagon-like peptide 1 (GLP-1) modulator such as an agonist (e.g., exendin-3 and exendin-4), liraglutide, albiglutide, exenatide (Byetta0), albiglutide, lixisenatide, dulaglutide, semaglutide, NN-9924,TTP-054, a protein tyrosine phosphatase-1B (PTP-1B) inhibitor (e.g., trodusquemine, hyrtiosal extract, and compounds disclosed by Zhang, S., et al., Drug Discovery Today, 12(9/10), 373-381 (2007)), SIRT-1 inhibitor (e.g., resveratrol, GSK2245840 or GSK184072), a dipeptidyl peptidease IV (DPP-IV) inhibitor (e.g., those in W02005116014, sitagliptin, vildagliptin, alogliptin, dutogliptin, linagliptin and saxagliptin), an insulin secreatagogue, a fatty acid oxidation inhibitor, an A2 antagonist, a c-jun amino-terminal kinase (JNK) inhibitor, glucokinase activators (GKa) such as those described in W02010103437, W02010103438, W02010013161, W02007122482, TTP-399, TTP-355, TTP-547, AZD1656, ARRY403, MK-0599, TAK-329, AZD5658 or GKM-001, insulin, an insulin mimetic, a glycogen phosphorylase inhibitor (e.g. GSK1362885), a VPAC2 receptor agonist, SGLT2 inhibitors, such as those described in E.C. Chao et al.
Nature Reviews Drug Discovery 9, 551-559 (July 2010) including dapagliflozin, canagliflozin, empagliflozin, tofogliflozin (CSG452), ASP-1941, THR1474, TS-071, ISIS388626 and LX4211 as well as those in W02010023594, a glucagon receptor modulator such as those described in Demong, D.E. et al. Annual Reports in Medicinal Chemistry 2008, 43, 119-137, GPR119 modulators, particularly agonists, such as those described in W02010140092, W02010128425, W02010128414, W02010106457, Jones, R.M. et al. in Medicinal Chemistry 2009, 44, 149-170 (e.g. MBX-2982, GSK1292263, APD597 and PSN821), FGF21 derivatives or analogs such as those described in Kharitonenkov, A. et al. et al., Current Opinion in Investigational Drugs 2009, 10(4)359-364, TGR5 (also termed GPBAR1) receptor modulators, particularly agonists, such as those described in Zhong, M., Current Topics in Medicinal Chemistry, 2010, 10(4), 386-396 and INT777, GPR40 agonists, such as those . .

described in Medina, J.C., Annual Reports in Medicinal Chemistry, 2008, 43, 75-85, including but not limited to TAK-875, GPR120 modulators, particularly agonists, high affinity nicotinic acid receptor (HM74A) activators, and SGLT1 inhibitors, such as GSK1614235. A further representative listing of anti-diabetic agents that may be combined with the compounds of the present invention can be found, for example, at page 28, line 35 through page 30, line 19 of W02011005611. Preferred anti-diabetic agents may include metformin and DPP-IV inhibitors (e.g., sitagliptin, vildagliptin, alogliptin, dutogliptin, linagliptin and saxagliptin). Other antidiabetic agents that might be used include inhibitors or modulators of carnitine palmitoyl transferase enzymes, inhibitors of fructose 1,6-diphosphatase, inhibitors of aldose reductase, mineralocorticoid receptor inhibitors, inhibitors of TORC2, inhibitors of CCR2 and/or CCR5, inhibitors of PKC isoforms (e.g. PKCa, PKC[3, PKCy), inhibitors of fatty acid synthetase, inhibitors of serine palmitoyl transferase, modulators of GPR81, GPR39, GPR43, GPR41, GPR105, Kv1.3, retinol binding protein 4, glucocorticoid receptor, somatostain receptors (e.g. SSTR1, SSTR2, SSTR3 and SSTR5), inhibitors or modulators of PDHK2 or PDHK4, inhibitors of MAP4K4, modulators of lL1 family including IL1beta, modulators of RXRalpha. In addition suitable anti-diabetic agents may include mechanisms listed by Carpino, P.A., Goodwin, B. Expert Opin. Ther. Pat, 2010, 20(12), 1627-51;
(v) an anti-platelet agent. It may be especially preferred to use the anti-platelet agents aspirin and clopidogrel.
The term anti-platelet agents (or platelet inhibitory agents), as used herein, denotes agents that inhibit platelet function, for example by inhibiting the aggregation, adhesion or granular secretion of platelets. Agents may include, but are not limited to, the various known non-steroidal anti-inflammatory drugs (NSAIDS) such as aspirin, ibuprofen, naproxen, sulindac, indomethacin, mefenamate, droxicam, diclofenac, sulfinpyrazone, piroxicam, and pharmaceutically acceptable salts or prodrugs thereof. Of the NSAIDS, aspirin (acetylsalicyclic acid or ASA) and COX-2 inhibitors such as celecoxib or piroxicam may be preferred. Other suitable platelet inhibitory agents may include Ilb/Illa antagonists (e.g., tirofiban, eptifibatide, and abciximab), thromboxane-A2-receptor antagonists (e.g., ifetroban), thromboxane-A2-synthetase inhibitors, PDE3 inhibitors (e.g., Pieta!, dipyridamole), and pharmaceutically acceptable salts or prodrugs thereof. The term anti-platelet agents (or platelet inhibitory agents), as used herein, is also intended to include ADP (adenosine diphosphate) receptor antagonists, which may preferably be antagonists of the purinergic receptors P2Y1 and P2Y12, and P2Y12 may be even more preferred. Preferred P2Y12 receptor antagonists may include ticagrelor, prasugrel, ticlopidine and clopidogrel, including pharmaceutically acceptable salts or prodrugs thereof. Clopidogrel may be an even more preferred agent. Ticlopidine and clopidogrel may also be preferred compounds since they are known to be gentle on the gastro-intestinal tract in use;
(vi) an anti-thrombin agent including inhibitors of the serine protease thrombin. By inhibiting thrombin, various thrombin-mediated processes, such as thrombin-mediated platelet activation (that is, for example, the aggregation of platelets, and/or the granular secretion of plasminogen activator inhibitor-1 and/or serotonin) and/or fibrin formation are disrupted. A number of thrombin inhibitors are known to one of skill in the art and these inhibitors may be used in combination with the present compounds. Such inhibitors may include, but are not limited to, boroarginine derivatives, boropeptides, heparins, hirudin, argatroban, and melagatran, including pharmaceutically acceptable salts and prodrugs thereof. Boroarginine derivatives and boropeptides that may be used include N-acetyl and peptide derivatives of boronic acid, such as C-terminal alpha-aminoboronic acid derivatives of lysine, ornithine, arginine, homoarginine and corresponding isothiouronium analogs thereof. The term hirudin, as used herein, includes suitable derivatives or analogs of hirudin, referred to herein as hirulogs, such as disulfatohirudin, which may be used;
(vii) a thrombolytics or fibrinolytic agents (or thrombolytics or fibrinolytics), including agents that lyse blood clots (thrombi). Suitable agents may include tissue plasminogen activator (natural or recombinant) and modified forms thereof, anistreplase, urokinase, streptokinase, tenecteplase (TNK), lanoteplase (nPA), factor Vila inhibitors, PAI-1 inhibitors (i.e., inactivators of tissue plasminogen activator inhibitors), alpha2-antiplasmin inhibitors, and anisoylated plasminogen streptokinase activator complex, including pharmaceutically acceptable salts or prodrugs thereof. The term anistreplase, as used herein, refers to anisoylated plasminogen streptokinase activator complex, as described, for example, in EP
028,489, the disclosure of which is hereby incorporated herein by reference herein. The term urokinase, as used herein, is intended to denote both dual and single chain urokinase, the latter also being referred to herein as prourokinase.
5 Examples of suitable anti-arrythmic agents may include: Class I agents (such as propafenone); Class II agents (such as metoprolol, atenolol, carvadiol and propranolol); Class III agents (such as sotalol, dofetilide, amiodarone, azimilide and ibutilide); Class IV agents (such as ditiazem and verapamil); K+ channel openers such as lAch inhibitors, and IKur inhibitors (e.g., compounds such as 10 those disclosed in W001/40231);
(viii) a factor Xa inhibitor. Suitable factor Xa inhibitors may include apixaban and rivaroxaban;
(ix) anti-coagulants, including heparins (e.g., unfractioned and low molecular weight heparins such as enoxaparin and dalteparin);
15 (x) other agents such as warfarin, unfractionated heparin, low molecular weight heparin, synthetic pentasaccharide, hirudin, argatrobanas, aspirin, ibuprofen, naproxen, sulindac, indomethacin, mefenamate, droxicam, diclofenac, sulfinpyrazone, piroxicam, ticlopidine, clopidogrel, tirofiban, eptifibatide, abciximab, melagatran, disulfatohirudin, tissue plasminogen activator, modified 20 tissue plasminogen activator, anistreplase, urokinase, and streptokinase;
(xi) a neuroinflammatory or a neurodegenerative agent. Examples of suitable neuroinflammatory and neurodegenerative agents may include antidepressants, antipsychotics, anti-pain agents, anti-Alzheimer's agents, and anti-anxiety agents. Examples of particular classes of antidepressants that may be used in 25 combination with the compounds for use in the present invention may include norepinephrine reuptake inhibitors, selective serotonin reuptake inhibitors (SSR15), NK-1 receptor antagonists, monoamine oxidase inhibitors (MA015), reversible inhibitors of monoamine oxidase (RIMAs), serotonin and noradrenaline reuptake inhibitors (SNRIs), corticotropin releasing factor (CRF) antagonists, 30 and atypical antidepressants. Suitable norepinephrine reuptake inhibitors may include tertiary amine tricyclics and secondary amine tricyclics. Examples of suitable tertiary amine tricyclics and secondary amine tricyclics may include amitriptyline, clomipramine, doxepin, imipramine, trimipramine, dothiepin, butriptyline, nortriptyline, protriptyline, amoxapine, desipramine and maprotiline.
Examples of suitable SSRIs may include fluoxetine, fluvoxamine, paroxetine, and sertraline.
Examples of monoamine oxidase inhibitors may include isocarboxazid, phenelzine, and tranylcyclopramine.
Examples of suitable reversible inhibitors of monoamine oxidase may include moclobemide.
Examples of suitable SNRIs of use in the present invention may include venlafaxine.
Examples of suitable atypical anti-depressants may include bupropion, lithium, trazodone and viloxazine. Examples of anti-Alzheimer's agents may include NMDA receptor antagonists such as memantine; and cholinesterase inhibitors such as donepezil and galantamine. Examples of suitable classes of anti-anxiety agents that may be used in combination with the compounds for use in the present invention may include benzodiazepines and serotonin 1A receptor (5-HT1A) agonists, and CRF antagonists. Suitable benzodiazepines may include alprazolam, chlordiazepoxide, clonazepam, chlorazepate, diazepam, lorazepam, oxazepam, and prazepam. Suitable 5-HT1A receptor agonists may include buspirone and ipsapirone. Suitable CRF
antagonists may include verucerfont. Suitable atypical antipsychotics may include paliperidone, ziprasidone, risperidone, aripiprazole, olanzapine, and quetiapine. Suitable nicotine acetylcholine agonists may include CP-601927 and varenicline. Anti-pain agents may include pregabalin, gabapentin, clonidine, neostigmine, baclofen, midazolam, ketamine and ziconotide;
(xii) an agent to reduce spasticity and spasms. Examples may include baclofen, tizanidine, benzodiazepines, dantrolene sodium, gabapentin and botulinum toxin;
(xiii) antioxidants. Examples may include myeloperoxidase inhibitors (such as AZD-3241), NOX4 and other NOX enzymes (such as GKT-137831), vitamin E, Coenzyme Q, and N-acetyl cysteine;
(xiv) a further BET-family bromodomain inhibitor;
(xv) a non-steroidal anti-inflammatory drugs (NSAIDs). Examples may include, non-selective COX1/2 inhibitors such as piroxicam, naproxen, flubiprofen, fenoprofen, ketoprofen, ibuprofen, etodolac (Lodine), mefanamic acid, sulindac, apazone, pyrazolones (such as phenylbutazone), salicylates (such as aspirin); selective COX2 inhibitors such as: celecoxib, rofecoxib, etoricoxib, valdecoxib, meloxicam;

(xvi) an immunomodulatory and/ or anti-inflammatory agents. Examples of suitable agents may include methotrexate, leflunomide, ciclesonide chloroquine, hydroxychloroquine, d-penicillamine, auranofin, sulfasalazine, sodium aurothiomalate, cyclosporine, azathioprine, cromolyn, hydroxycarbamide, retinoids, fumarates (such as monomethyl and dimethyl fumarate), glatiramer acetate, mitoxantrone, teriflunomide, suplatast tosilate, mycophenolate mofetil and cyclophosphamide, laquinimod, voclosporin, PUR-118, AMG 357, AMG 811, BCT197;
(xvii) a Nrf2 pathway activators. Examples of suitable agents may include fumarates, sulfurophane and bardoxolone methyl; and (xviii) an interferon. Suitable interferons may include interferon beta-1a and interferon beta-1b.
In one embodiment the additional therapeutic agent may be an agent useful for treating Friedreich's ataxia; treating a condition or a disease associated with decreased expression of frataxin or a decreased level of frataxin; regulating the expression of frataxin in a cell; and / or for increasing the expression of frataxin in a cell. Such agents may include, but are not limited to, those selected from the group consisting of carbanylated erythropoietin; interferon gamma1b; methylprednisolone;
idebenone;
quinone antioxidants such as A00001 ¨ alpha tocopherolquinone and EP1-743;
iron chlating agents cuah as deferiprone; Nrf2 enhancers such as RTA-408; acetyl-1-carnitine; pioglitazone; di-deutero synthetic analogues of linoleic acid ethyl ester such as RT001; ginkgo biloba extract such as EGb761; free radical scavengers such as indole-3-propionic acid or EP20629; nicotinamide; and riluzole.
Inasmuch as it may be desirable to use a combination of active compounds, for the pharmaceutical uses of the present invention, it is within the scope of the present invention that two or more pharmaceutical compositions may conveniently be combined in the form of a kit suitable for co-administration of the compositions.
BIOLOGICAL EXAMPLES
Test Compound Source Test compounds JQ-1(+), JQ-1(-), and I-BET-762 were obtained from Selleck Chemicals, Houston, Texas, USA. Other test compounds were obtained from Pfizer's library, which compounds had been prepared in accordance with methods from the art.
For example RVX-208 was prepared in accordance with the method set out in W02008/092231; I-BET-151 was prepared in accordance with the method set out in W02011054843; N-(2-hydroxy-3-methylquinolin-6-yl)piperidine-1-sulfonamide was prepared in accordance with the method set out in W02013/027168; N-[4-(2,4-difluorophenoxy)-3-(6-methyl-7-oxo-6,7-dihydro-1H-pyrrolo[2,3-c]pyrid in-yl)phenyllethanesulfonamide was prepared in accordance with the method set out in WO 2013/097052 or WO 2013/097601; and N-{6-[methyl(propanoyl)amino]-3-oxo-4-[(1S)-1-phenylpropy1]-3,4-dihydropyrido[2,3-b]pyrazin-2-y1}-beta-alanine was prepared in accordance with the method set out in US provisional patent application serial number 62/181281, which was filed on 18th June 2015.
All compounds were reconstituted in DMSO (Sigma) prior to use.
Example 1 - Bromodomain 4 (BRD4) - Fluorescence Polarization (FP) Binding Assay The biological activity of BET-family bromodomain inhibitors useful for the present invention may be measured in the BRD4 Fluorescence polarization (FP) competition binding assay where the compounds can be screened for their activity at BRD4 BD1, BRD4 BD2 or both BRD4 BD1 and BD2.
His-tagged BRD4 BD1 (44-160) and PFI-411FP (Cy5-labeled FP probe) were prepared as previously described (Picaud S et al PFI-1, a highly selective protein interaction inhibitor, targeting BET Bromodomains. Cancer Res. (2013) 73: 3336-and Wu, J et al Design and chemoproteomic functional characterization of a chemical probe targeted to bromodomains of BET-family proteins. Med. Chem. Commun.
2014, vol 5m p1871-1878, incorporated by reference herein). His-tagged BRD4 BD2 (342-460) was purchased from BPS Bioscience. All assay components were diluted in mM HEPES pH 7.4 containing 0.08% bovine serum albumin (assay buffer). To start the assay, 8 pL of the indicated BRD4 was added to each well of a low volume 384-well black flat bottom microtiter plate (Corning 3820) containing 4 pl of various concentrations of test compound (each plate also contained positive and negative control wells to define the upper and lower limits of the assay signal). After addition of BRD4, the assay plate was incubated at room temperature (RT, 20 C). After 15 minutes, 4 pL of PFI-411FP was added to each well and the assay plate was placed in the dark at RT. The final assay concentration (FAC) of PFI-411FP was 2 nM, the FAC
of BRD4 BD1 and BRD4 BD2 was 50 nM, the FAC of test compound ranged from 120 to 0.0012 pM and the FAC of DMSO was 0.4%. After 60 min, polarization values were measured with an Envision 2103 multilabel reader (Perkin Elmer) using a Cy5 dual enhanced mirror and excitation at 620 nm and emission at 688 nm. The percent (/0) effect was calculated for each concentration of test compound and was relative to the amount of polarization signal produced by the positive and negative control wells contained within each assay plate. The concentrations and % effect values for test compounds were plotted versus each other with a proprietary curve fitting program using a four-parameter logistic dose response equation and the concentration of compound required for 50% effect (IC50) was determined. The Ki values of competitive inhibitors are calculated using the equation described by Nikolovska-Coleska et al.
(Development and optimization of a binding assay for the XIAP BIR3 domain using fluorescence polarization. Analytical Biochemistry (2004) 332: 261-273, incorporated by reference herein).
Certain compounds were tested in the Fluorescence Polarization assay described above:
Compound Name Structure BRD4 BD1 binding IC 50 (1.1M) N-[4-(2,4-difluorophenoxy)-0.054 /st' 3-(6-methyl-7-oxo-6,7- / \o dihydro-1H-pyrrolo[2,3- 0 c]pyridin- 4-yl)phenyl]ethanesulfonamide N-{6- H 0.062 HO.N 0 [methyl(propanoyl)amino]-3- 0 Me oxo-4-[(1S)-1-phenylpropy1]- N N
3,4-dihydropyrido[2,3-Me b]pyrazin-2-y1}-beta-alanine al Me N-(2-hydroxy-3- 0.070 methylquinolin-6-yl)piperidine-1-sulfonamide \O
HO N
I-BET-151 0.059 NH

N\0 I-BET-762 0.138 /
0\ 4, CI
RVX-208 4.413 NH

JQ-1(+) 0.051 S N
CI

, = PC72246 JQ-1(-) N-N 29,300 S N
CI
Example 2 - Frataxin levels in Friedreich's Ataxia patient derived lymphoblasts are increased on treatment of with JQ-1(+) and I-BET-762 Friedreich's Ataxia lymphoblasts GM15850 (FXN alleles carrying ¨ 650 and 1030 GAA
repeats) were obtained from NIGMS Human Genetic Cell Repository at the Coriell Institute for Medical Research and cultured in complete medium containing media supplemented with 10% Fetal bovine serum (Invitrogen) and 20mM Hepes buffer (Invitrogen). Prior to treatment with a test compound, 500,000 cells were seeded in 900u1 complete medium were seeded per well in a 12 well plate, and subsequently treated with the test compound (10mM in DMSO) which had been diluted in 100u1 optimem (lnvitrogen) to achieve a final concentration of test compound of 5.0pM; 0.5pM
or 0.05pM. Cells were incubated in a 37 C incubator (5% CO2) for 72h before harvesting. For harvesting, cells were collected by centrifugation at 500xg for 5 mins, then washed once in 1m1 phosphate buffered saline (PBS) before lysis in NP40 lysis buffer (0.5%NP40+50mM Tris pH7.5, 150mM NaCl supplemented with protease and phosphatase inhibitors from Roche) for 30 mins on ice. Lysates were finally cleared by centrifugation at max speed in a refrigerated microcentrigue for 10 mins.
Protein concentration was determined using the Protein 660 reagent (Pierce) (results shown in Figure 2, total protein concentration in mg/ml) and lysates were prepared for polyacrylamide gel electrophoresis (PAGE) analysis by dilution in lysis buffer supplemented with LDS sample buffer (Invitrogen) containing 10%
dithiothreitol.
Samples (5-bug/lane) were then denatured by heating at 100C for 5 mins, loaded on a 4-12% BisTris Nupage gel (Invitrogen) and resolved in MES running buffer (Invitrogen) supplemented with Nupage antioxidant (Invitrogen). Proteins were transferred to a nitrocellulose membrane (0.2um from Biorad) using the Trans-Blot Turbo (Biorad).

. = PC72246 Membrane was blocked in 5% milk in Tris buffered saline supplemented with 0.05%
Tween 20 (TBST) for 1h and left in primary antibody anti-FXN (ab110328 from Abcam;
1:1000 dilution) diluted in blocking buffer overnight at 40. Membrane was then washed three times in TBST and incubated in blocking buffer containing secondary anti-mouse antibody conjugated to HRP (1:7500 anti-mouse IgG HRP) for 30 mins and then subjected to four washes in TBST. Similarly, anti-13-actin (Cell Signaling;
1:12000 dilution) western blotting was used as lane loading control. Signal was finally developed in ECL prime HRP substrate (GE) for 5 mins and membranes were exposed to Biomax film (Kodak) before processing in a film developer. Results (shown in Figure 1) from both a short and long exposure of the film are reported in Figure 1.
Bands corresponding to both intermediate frataxin (i) and mature frataxin (m) are indicated in Figure 1.
The results of these experiments (Figure 1) demonstrate that treatment of GM15850 cell line with BET-family bromodomain inhibitor JQ-1(+) or IBET762 increased the levels of both intermediate and mature frataxin.
Additionally treatment with the inactive enantiomer JQ-1(-) had no significant effect on frataxin levels suggesting that the increase in frataxin is as a result of the inhibition of the BET-family bromodomain.
However, as shown in Figure 2, treatment of GM15850 cell line with BET-family bromodomain inhibitor JQ-1(+) or I-BET-762 induced cytotoxic effects as seen by reduction in the concentration of total cellular protein. This cytotoxic effect was further investigated, see Example 4 below.
Example 3 - The dose dependent effect of JQ-1(+) on frataxin levels in Friedreich's Ataxia patient derived lymphoblasts was measured GM15850 cells (Coriell) were subjected to a dose-response treatment with JQ-1(+) in duplicate at concentrations that yield low cytotoxicity in duplicate, as described above in. The concentration of JQ-1(+) tested were 6.25nM, 12.5nM, 25nM and 50nM, chosen to be minimally cytotoxic level. As a reference for levels of frataxin in healthy cells, AG14725 lymphoblasts (Coriell) were treated, again in duplicate, with DMSO
alongside.
Lysates from both cell lines were subsequently analyzed by western blotting, as previously described in Example 2. Bands corresponding to both intermediate frataxin (i) and mature frataxin (m) are indicated in Figure 3. The results in Figure 3 show that ' treatment of GM15850 cells with JQ-1(+) at 25nM and 50nM concentration resulted in an increased in both intermediate and mature frataxin.
Bands corresponding to mature frataxin protein were quantified using Pixel Densitometry using the Image StudioLite (version 4.0) software from Licor. The results are shown in Figure 4. These results confirm that treatment of GM15850 cells with JQ-1(+) at 25nM and 50nM concentration resulted in an increased in both intermediate and mature frataxin and the level of increase was quantified to be approximately 1.5 fold increase.
Example 4 - The cytotoxic effect of JQ-1(+) on healthy AG14725 lymphoblasts and Friedreich's Ataxia GM15850 lymphoblasts was measured This experiment was conducted to confirm that low levels of cellular frataxin was not sensitizing the test cell line to the cytotoxic effects induced by treatment with the BET-family bromodomain test compound, and that such cells were not being selectively killed to enrich for higher expressors. A dose response treatment of GM15850 lymphoblasts (Coriell) and AG14725 lymphoblasts (Coriell) was conducted with JQ-1(+) to determine the Lethal Dose 50 (LD50, concentration of JQ-1(+) that yields 50% cell killing). The lymphoblasts were seeded in a 96 well plate, (40,000 cells per well in 180pL
of RPMI1640 media supplemented with 10% Fetal bovine serum (Invitrogen) and 20mM
Hepes buffer (lnvitrogen)) and were incubated in a 37 C incubator (5% 002) for 72h before harvesting with varying concentrations of the test compound (duplicate treatment per condition). The test compound was prepared by dilution of a 10mM stock solution in DMSO with optimen (lnvitrogen) to the desired test concentration. Finally, cell viability was determined using the CellTiter Glo luminescent assay (Promega) according to the protocol provided by the manufacturer. The results are shown in Figure 5. The results indicate that the LD50 displayed for both cell lines treated, namely the healthy AG14725 and FRDA GM15850, was approximately 100nM and was comparable for both cell lines. These results suggest that lymphoblasts containing reduced level of frataxin are not sensitized to killing by BET inhibitors.
Example 5 - Frataxin levels in Friedreich's Ataxia patient derived lymphoblasts are increased on treatment of with a variety of different BET-family bromodomain inhibitor scaffolds.

We further examined the effect on frataxin levels of a variety of small molecule compounds, known to have BET-family bromodomain inhibitory activity, and with a variety of chemical scaffolds. GM15850 lymphoblasts were prepared according to the directions provided in Example 2 above and treated with a 1pM solution of the test compound for 72 hours, as described previously. The test compound was prepared by dilution of a 10mM stock solution in DMSO with optimen (Invitrogen) to the desired test concentration. The compounds tested were:
Compound Compound Structure Nurnber Cmp1 N-[4-(2,4-difluorophenoxy)-3-(6-F
40) methy1-7-oxo-6,7-dihydro-1H-pyrrolo[2,3-c]pyridin- 4- 0 yl)phenyliethanesulfonamide \

Cmp2 N-{6-[methyl(propanoyDamino]-3-oxo-4-[(1S)-1-phenylpropyI]-3,4- 0 -J)c, dihydropyrido[2,3-b]pyrazin-2-yI}-0 N N N Me beta-alanine Me 101 Me Cm p3 N-(2-hydroxy-3-methylquinolin-6-yl)piperidine-1-sulfonamide H

HO
Cmp4 I-BET151 N

Cm p5 1-BET762 N¨N
N
N
ofh 411, CI
Cmp6 RVX-208 NH

The levels of intermediate and mature frataxin in the cells were assessed by Western Blotting using the same protocol as described in Example 2 above. Lysates were also subjected to Western Blotting again using the same protocol with an antibody raised 5 against mitochondria! aconitase (mAco2); the antibody was obtained from Abcam (ab129105). The results (shown in Figure 6) demonstrate that the level of both intermediate and mature frataxin was increased for all compounds tested, although the increase seen with Cmpd1 was reduced when compared to the other compounds tested which, without wishing to be bound by theory, may possibly be a result of the reduced 10 solubility of Cmpd1 in the test medium. Our data also shows that the levels of mAco2 remain unchanged by treatment with the test compound BET-family bromodomain inhibitors indicating that these compounds do not cause a global increase in levels of all mitochondrial proteins such as mAco2 and thus confirming that the effect seen was specific to frataxin.
Example 6 - Bromodomain containing 4 protein (BRD4) knockdown and inhibition in Friedreich's Ataxia fibroblasts increases frataxin levels Further experiments were conducted to validate that knockdown of the well characterized BET-family bromodomain protein BRD4 can reproduce the effect observed by chemical inhibition of BET, and to examine if this inhibition would be = PC72246 observed in other non-lymphoblast patient derived cells. GM03665 cells (Coriell; ¨700 and 400 GAA repeats) were seeded at 100,000 cells/well in a 6-well plate in 2mL of DMEM media (lnvitrogen) supplemented with 10% Fetal bovine serum (Invitrogen) and 20mM Hepes buffer (Invitrogen)). The cells were reverse transfected using transfection reagent Dharmafect I (Dharmacon) following manufacturer's recommendation mixed with the following siRNA duplex at a concentration of 25nM in optimen (Invitrogen).
Target Source siRNA duplex format Oligonucelotide Sequence catalog #
BRD4 Dharmacon smartpool (4 per) L-004937-00-0005 Additionally, one well was treated with 1pM JQ-1(+) prepared by dilution of a 10mM
stock solution in DMSO with optimen (Invitrogen). All the cells were incubated for 72hrs following transfection or treatment, lysed and analysed by Western Blotting as previously described in Example 2 above. The results are shown in Figure 7.
These results show that knockdown of BRD4 expression using the BRD4 siRNA smartpool resulted in an increase in both intermediate and mature frataxin levels.
Furthermore the treatment with JQ-1(+) (1pM) resulted in an increase intermediate and mature frataxin levels demonstrating that the earlier results seen are not limited to the Friedreich's Ataxia lymphoblasts.
Example 7 - Knockdown of BET proteins upregulates FXN
Further experiments were conducted to validate that knockdown of all BET-family bromodomains can reproduce the effect observed by chemical inhibition of BET, and to examine if this inhibition would be observed in other non-lymphoblast patient derived cells. GM03665 cells (Coriell; ¨700 and 400 GAA repeats) were seeded at 100,000 cells/well in a 6-well plate in 2mL of DMEM media (Invitrogen) supplemented with 10%
Fetal bovine serum (Invitrogen) and 20mM Hepes buffer (Invitrogen)). The cells were reverse transfected using transfection reagent Dharmafect I (Dharmacon) following manufacturer's recommendation mixed with the following siRNA duplex at a concentration of 25nM in optimen (Invitrogen).
Target Source siRNA duplex format Oligonucelotide Sequence catalog #
BRD2 Dharmacon smartpool (4 per) L-004935-00-0005 BRD3 Dharmacon smartpool (4 per) L-004936-00-0005 BRD4 Dharmacon smartpool (4 per) L-004937-00-0005 BRD4 Sigma isofornn A (1 per) Sequences as disclosed in Floyd et at.
isoform B (1 per) Nature, 2013, 498(7453), 246-50.
BRDT Dharmacon smartpool (4 per) L-004938-00-0005 All the cells were incubated for 72hrs following transfection or treatment, lysed and analysed by Western Blotting as previously described in Example 2 above. The results are shown in Figure 8. These results show that knockdown of all BRD subtypes expression using the siRNA snnartpool resulted in an increase in both intermediate and mature frataxin levels.
Variations, modifications, and other implementations of what is described herein will occur to those skilled in the art without departing from the spirit and the essential characteristics of the present teachings. Accordingly, the scope of the present teachings is to be defined not by the preceding illustrative description but instead by the following claims, and all changes that come within the meaning and range of equivalency of the claims are intended to be embraced therein.
Each of the printed publications, including but not limited to patents, patent applications, books, technical papers, trade publications and journal articles described or referenced in this specification are herein incorporated by reference in their entirety and for all purposes.

Claims (29)

1. A method of increasing the level of frataxin in a cell, comprising contacting the cell with an effective amount of a BET- family bromodomain inhibitor, or a pharmaceutically acceptable salt thereof.
2. A method of increasing the expression of frataxin in a cell, comprising contacting the cell with an effective amount of a BET- family bromodomain inhibitor, or a pharmaceutically acceptable salt thereof.
3. Use of an effective amount of a BET- family bromodomain inhibitor, or a pharmaceutically acceptable salt thereof, for increasing the level of frataxin.
4. Use of an effective amount of a BET- family bromodomain inhibitor, or a pharmaceutically acceptable salt thereof, for increasing the expression of frataxin.
5. Use according to Claim 3 or 4 wherein the BET- family bromodomain inhibitor is a BRD-4 inhibitor, or a pharmaceutically acceptable salt thereof.
6. Use of a therapeutically effective amount of a BET- family bromodomain inhibitor, or a pharmaceutically acceptable salt thereof, for treating a condition, disease or disorder, or the symptoms associated with the condition, disease or disorder, which condition, disease or disorder is associated with decreased frataxin expression, in a subject in need thereof.
7. Use of a therapeutically effective amount of a BET- family bromodomain inhibitor, or a pharmaceutically acceptable salt thereof, for treating a condition, disease or disorder, or the symptoms associated with the condition, disease or disorder, which condition, disease or disorder is associated with a decreased level of frataxin, in a subject in need thereof.
8. Use according to Claim 6 or 7 wherein the BET- family bromodomain inhibitor is a BRD-4 inhibitor, or a pharmaceutically acceptable salt thereof.
9. Use according to Claim 3, 4, 6 or 7 wherein the BET- family bromodomain inhibitor is selected from the group consisting of I-BET-762;
JQ-1;
JQ-1(+);
CP1-203;
OTX-015;
GW-841819X;
CP-0610;
CPI-232;
BET-BAY-002;
I-BET-151;
RVX-208;
I-BET-726;
SRX-2523;
N-(2-hydroxy-3-methylquinolin-6-yl)piperidine-1-sulfonamide;
N-[4-(2,4-difluorophenoxy)-3-(6-methyl-7-oxo-6,7-dihydro-1H-pyrrolo[2,3-c]pyridin- 4-yl)phenyl]ethanesulfonamide; and N-{6-[methyl(propanoyl)amino]-3-oxo-4-[(1S)-1-phenylpropyl]-3,4-dihydropyrido[2,3 b]pyrazin-2-yl}-beta-alanine, or a pharmaceutically acceptable salt thereof.
10. Use according to Claim 9, wherein the BET- family bromodomain inhibitor is I-BET-762, or a pharmaceutically acceptable salt thereof.
11. Use according to Claim 9, wherein the BET- family bromodomain inhibitor is JQ-1(+), or a pharmaceutically acceptable salt thereof.
12. Use according to Claim 9, wherein the BET- family bromodomain inhibitor is I-BET-151, or a pharmaceutically acceptable salt thereof.
13. Use according to Claim 9, wherein the BET- family bromodomain inhibitor is RVX-208, or a pharmaceutically acceptable salt thereof.
14. Use according to Claim 9, wherein the BET- family bromodomain inhibitor is N-(2-hydroxy-3-methylquinolin-6-yl)piperidine-1-sulfonamide, or a pharmaceutically acceptable salt therof.
15. Use according to Claim 9, wherein the BET-family bromodomain inhibitor is N-[4-(2,4-difluorophenoxy)-3-(6-methyl-7-oxo-6,7-dihydro-1H-pyrrolo[2,3-c]pyridin-yl)phenyl]ethanesulfonamide, or a pharmaceutically acceptable salt thereof.
16. Use according to Claim 9, wherein the BET-family bromodomain inhibitor is N-{6-[methyl(propanoyl)amino]-3-oxo-4-[(1S)-1-phenylpropyl]-3,4-dihydropyrido[2,3 b]pyrazin-2-yl}-beta-alanine, or a pharmaceutically acceptable salt thereof.
17. Use of a therapeutically effective amount of a BET- family bromodomain inhibitor, or a pharmaceutically acceptable salt thereof, for treating Friedreich's ataxia in a mammal in need thereof.
18. Use according to Clam 17 wherein the subject is a patient diagnosed with Friedreich's ataxia with retained reflexes.
19. Use according to Claim 17 wherein the subject is a patient diagnosed with late onset Friedreich's ataxia.
20. Use according to any one of Claims 17 to 19 wherein the subject is asymptomatic.
21. Use according to any one of Claims 17 to 19 wherein the subject is a human.
22. Use according to any one of Claims 17 to 19 wherein the BET- family bromodomain inhibitor is selected from the group consisting of I-BET-762;
JQ-1;
JQ-1(+);
CPI-203;
OTX-015;
GW-841819X;
CP-0610;
CPI-232;
BET-BAY-002;

I-BET-151;
RVX-208;
I-BET-726;
SRX-2523;
N-(2-hydroxy-3-methylquinolin-6-yl)piperidine-1-sulfonamide;
N-[4-(2,4-difluorophenoxy)-3-(6-methyl-7-oxo-6,7-dihydro-1H-pyrrolo[2,3-c]pyridin- 4-yl)phenyl]ethanesulfonamide; and N-{6-[methyl(propanoyl)amino]-3-oxo-4-[(1S)-1-phenylpropyl]-3,4-dihydropyrido[2,3 b]pyrazin-2-yl}-beta-alanine or a pharmaceutically acceptable salt thereof.
23. Use according to Claim 22, wherein the BET- family bromodomain inhibitor is I-BET-762, or a pharmaceutically acceptable salt thereof.
24. Use according to Claim 22, wherein the BET- family bromodomain inhibitor is JQ-1(+), or a pharmaceutically acceptable salt thereof.
25. Use according to Claim 22, wherein the BET- family bromodomain inhibitor is I-BET-151, or a pharmaceutically acceptable salt thereof.
26. Use according to Claim 22, wherein the BET- family bromodomain inhibitor is RVX-208, or a pharmaceutically acceptable salt thereof.
27. Use according to Claim 22, wherein the BET- family bromodomain inhibitor is N-(2-hydroxy-3-methylquinolin-6-yl)piperidine-1-sulfonamide, or a pharmaceutically acceptable salt therof.
28. Use according to Claim 22, wherein the BET-family bromodomain inhibitor is N-[4-(2,4-d ifluorophenoxy)-3-(6-methyl-7-oxo-6,7-dihydro-1H-pyrrolo[2,3-c]pyridin-yl)phenyl]ethanesulfonamide, or a pharmaceutically acceptable salt thereof.
29. Use according to Claim 22, wherein the BET-family bromodomain inhibitor is N-{6-[methyl(propanoyl)amino]-3-oxo-4-[(1S)-1-phenylpropyl]-3,4-dihydropyrido[2,3 b]pyrazin-2-yl}-beta-alanine, or a pharmaceutically acceptable salt thereof.
CA2940742A 2015-09-03 2016-08-31 Regulators of frataxin Abandoned CA2940742A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US201562213929P 2015-09-03 2015-09-03
US62/213,929 2015-09-03

Publications (1)

Publication Number Publication Date
CA2940742A1 true CA2940742A1 (en) 2017-03-03

Family

ID=56851654

Family Applications (1)

Application Number Title Priority Date Filing Date
CA2940742A Abandoned CA2940742A1 (en) 2015-09-03 2016-08-31 Regulators of frataxin

Country Status (2)

Country Link
CA (1) CA2940742A1 (en)
WO (1) WO2017037567A1 (en)

Families Citing this family (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
PT2118074E (en) 2007-02-01 2014-03-20 Resverlogix Corp Compounds for the prevention and treatment of cardiovascular diseases
WO2010106436A2 (en) 2009-03-18 2010-09-23 Resverlogix Corp. Novel anti-inflammatory agents
CA3019342A1 (en) 2016-03-30 2017-10-05 Wisconsin Alumni Research Foundation Methods and compositions for modulating frataxin expression
US11759533B2 (en) 2017-03-29 2023-09-19 Wisconsin Alumni Research Foundation Methods and compositions for modulating gene expression
CN109384784B (en) * 2017-08-10 2021-01-12 浙江海正药业股份有限公司 Sulfonamide derivative, preparation method thereof and application thereof in medicine
MX2020010425A (en) * 2018-04-04 2021-03-09 Retrotope Inc Isotopically modified composition and therapeutic uses thereof.
CA3117322A1 (en) * 2018-10-23 2020-04-30 Wisconsin Alumni Research Foundation Next generation synthetic transcription factors
CN113881683B (en) * 2021-10-14 2023-09-22 山西大学 Migratory locust zen gene dsRNA and application thereof

Family Cites Families (26)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5612359A (en) 1994-08-26 1997-03-18 Bristol-Myers Squibb Company Substituted biphenyl isoxazole sulfonamides
TW536540B (en) 1997-01-30 2003-06-11 Bristol Myers Squibb Co Endothelin antagonists: N-[[2'-[[(4,5-dimethyl-3-isoxazolyl)amino]sulfonyl]-4-(2-oxazolyl)[1,1'-biphenyl]-2-yl]methyl]-N,3,3-trimethylbutanamide and N-(4,5-dimethyl-3-isoxazolyl)-2'-[(3,3-dimethyl-2-oxo-1-pyrrolidinyl)methyl]-4'-(2-oxazolyl)[1,1'-biphe
CA2336714A1 (en) 1998-07-06 2000-01-13 Bristol-Myers Squibb Company Biphenyl sulfonamides as dual angiotensin endothelin receptor antagonists
MY125533A (en) 1999-12-06 2006-08-30 Bristol Myers Squibb Co Heterocyclic dihydropyrimidine compounds
EA200400980A1 (en) 2002-02-27 2005-02-24 Пфайзер Продактс Инк. ACC INHIBITORS
EP1753748B1 (en) 2004-05-12 2009-07-29 Pfizer Products Inc. Proline derivatives and their use as dipeptidyl peptidase iv inhibitors
EP1951656B1 (en) * 2005-11-11 2015-08-05 The Scripps Research Institute Histone deacetylase inhibitors as therapeutics for neurological diseases
WO2007122482A1 (en) 2006-04-20 2007-11-01 Pfizer Products Inc. Fused phenyl amido heterocyclic compounds for the prevention and treatment of glucokinase-mediated diseases
CA2670422C (en) 2006-11-29 2011-09-06 Pfizer Products Inc. Spiroketone acetyl-coa carboxylase inhibitors
US20090036425A1 (en) 2007-08-02 2009-02-05 Pfizer Inc Substituted bicyclolactam compounds
CA2724603A1 (en) 2008-05-28 2009-12-03 Pfizer Inc. Pyrazolospiroketone acetyl-coa carboxylase inhibitors
ES2545231T3 (en) 2008-05-28 2015-09-09 Pfizer Inc. Pyrazoloespirocetone acetyl-CoA carboxylase inhibitors
EP2318370A1 (en) 2008-07-29 2011-05-11 Pfizer Inc. Fluorinated heteroaryls
CN103497199B (en) 2008-08-28 2015-04-29 辉瑞大药厂 Dioxa-bicyclo[3.2.1]octane-2, 3, 4-triol derivatives
TW201038580A (en) 2009-02-02 2010-11-01 Pfizer 4-amino-5-oxo-7,8-dihydropyrimido[5,4-f][1,4]oxazepin-6(5H)-yl)phenyl derivatives
CN102388038B (en) 2009-03-11 2014-04-23 辉瑞大药厂 Benzofuranyl derivatives used as glucokinase inhibitors
EP2406230A1 (en) 2009-03-11 2012-01-18 Pfizer Inc. Substituted indazole amides and their use as glucokinase activators
EP2408780A2 (en) 2009-03-20 2012-01-25 Pfizer Inc. 3-oxa-7-azabicycloý3.3.1¨nonanes
EP2427448A1 (en) 2009-05-08 2012-03-14 Pfizer Inc. Gpr 119 modulators
JP2012526097A (en) 2009-05-08 2012-10-25 ファイザー・インク GPR119 regulator
EA020106B1 (en) 2009-06-05 2014-08-29 Пфайзер Инк. L-(piperidin-4-yl)pyrazole derivatives as gpr119 modulators
WO2011005611A1 (en) 2009-07-09 2011-01-13 Merck Sharp & Dohme Corp. Neuromedin u receptor agonists and uses thereof
GB0919426D0 (en) * 2009-11-05 2009-12-23 Glaxosmithkline Llc Novel compounds
AU2011252808B2 (en) * 2010-05-14 2015-05-14 Dana-Farber Cancer Institute, Inc. Compositions and methods for treating neoplasia, inflammatory disease and other disorders
AR084070A1 (en) * 2010-12-02 2013-04-17 Constellation Pharmaceuticals Inc BROMODOMINIUM INHIBITORS AND USES OF THE SAME
WO2015023938A1 (en) * 2013-08-16 2015-02-19 Rana Therapeutics, Inc. Epigenetic regulators of frataxin

Also Published As

Publication number Publication date
WO2017037567A1 (en) 2017-03-09

Similar Documents

Publication Publication Date Title
CA2940742A1 (en) Regulators of frataxin
US20230165846A1 (en) Heterocyclic glp-1 agonists
US10308615B2 (en) Heterocyclic compounds as inhibitors of Vanin-1 enzyme
JP6526023B2 (en) Pyridines and pyrimidines and their use
EP2493313B1 (en) Kinase inhibitors
JP2023524493A (en) heterocyclic GLP-1 agonists
CN109574993B (en) Substituted pyrimidinylpiperazine compounds and uses thereof
EP2841421B1 (en) Amide derivatives as ttx-s blockers
US8999974B2 (en) Acyl piperazine derivatives as TTX-S blockers
KR20220156535A (en) Heterocyclic GLP-1 agonists
RU2477724C2 (en) Mdm2 and p53 interaction inhibitors
ES2645984T3 (en) Pyrrolopyridinone derivatives as TTX-S blockers
KR20120123652A (en) New ccr2 receptor antagonists and uses thereof
WO2012028233A1 (en) Imidazo[4,5-c]quinolines as dna-pk inhibitors
WO2012171863A1 (en) New selective ccr2 antagonists
JP2020520381A (en) Pyrrolopyridine derivative compound, method for producing the same, and pharmaceutical composition containing the same as an active ingredient for preventing or treating a protein kinase-related disease
WO2020121263A1 (en) Triazolopyridin-3-ones or their salts and pharmaceutical compositions comprising the same
US20230000847A1 (en) Compounds that modulates ampa receptor function
WO2019133445A1 (en) Aminothiazoles as inhibitors of vanin-1
US20160368919A1 (en) Novel pyridine pyrazinones as bet-family bromodomain inhibitors
US7211574B2 (en) Selective D1/D5 receptor antagonists for the treatment of obesity and CNS disorders
EA042282B1 (en) KAPPA OPIOID RECEPTOR ANTAGONISTS AND RELATED PRODUCTS AND METHODS
TW201139440A (en) Compounds for treating neurodegenerative diseases

Legal Events

Date Code Title Description
FZDE Dead

Effective date: 20190903